WO2014110205A1 - Methods and compositions for the production of sirnas - Google Patents

Methods and compositions for the production of sirnas Download PDF

Info

Publication number
WO2014110205A1
WO2014110205A1 PCT/US2014/010784 US2014010784W WO2014110205A1 WO 2014110205 A1 WO2014110205 A1 WO 2014110205A1 US 2014010784 W US2014010784 W US 2014010784W WO 2014110205 A1 WO2014110205 A1 WO 2014110205A1
Authority
WO
WIPO (PCT)
Prior art keywords
sirna
polypeptide
sirnas
dsrna
kit
Prior art date
Application number
PCT/US2014/010784
Other languages
French (fr)
Inventor
Judy Lieberman
Linfeng HUANG
Original Assignee
Children's Medical Center Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Children's Medical Center Corporation filed Critical Children's Medical Center Corporation
Priority to US14/758,924 priority Critical patent/US9840703B2/en
Publication of WO2014110205A1 publication Critical patent/WO2014110205A1/en
Priority to US15/804,258 priority patent/US10508276B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • HELECTRICITY
    • H05ELECTRIC TECHNIQUES NOT OTHERWISE PROVIDED FOR
    • H05KPRINTED CIRCUITS; CASINGS OR CONSTRUCTIONAL DETAILS OF ELECTRIC APPARATUS; MANUFACTURE OF ASSEMBLAGES OF ELECTRICAL COMPONENTS
    • H05K999/00PRINTED CIRCUITS; CASINGS OR CONSTRUCTIONAL DETAILS OF ELECTRIC APPARATUS; MANUFACTURE OF ASSEMBLAGES OF ELECTRICAL COMPONENTS dummy group
    • H05K999/99PRINTED CIRCUITS; CASINGS OR CONSTRUCTIONAL DETAILS OF ELECTRIC APPARATUS; MANUFACTURE OF ASSEMBLAGES OF ELECTRICAL COMPONENTS dummy group dummy group
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/50Biochemical production, i.e. in a transformed host cell
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the technology described herein relates to methods of producing siRNAs in vivo in bacterial cells.
  • RNA interference (RNAi) by double-stranded (ds) small interfering RNAs (siRNA) suppresses gene expression by inducing the degradation of mRNAs bearing complementary sequences (Fire, A. et al. Nature 1998 391 :806-811 ; Hamilton, A.J. & Baulcombe, D.C. Science 1999 286:950- 952).
  • Transfection of synthetic siRNAs into eukaryotic cells to silence genes has become an indispensable tool to investigate gene function, and siRNA-based therapy is being developed to knockdown genes implicated in disease (Elbashir, S.M. et al. Nature 2001, 411 :494-8; Caplen, N.J., et al. Proc Natl Acad Sci USA 2001 98:97427; Rettig, G.R. & Behlke, M.A. Mol Ther 2012 20:483- 512). More efficient ways to produce siRNAs are desired.
  • the technology described herein is directed to methods and compositions relating to the production of active siRNAs generated in vivo, e.g. in bacterial cells.
  • a bacterial cell comprising a siRNA-binding polypeptide and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • the siRNA-binding polypeptide comprises a purification tag.
  • the siRNA-binding polypeptide is encoded by a nucleic acid.
  • the siRNA-binding polypeptide is selected from the group consisting of: pi 9 polypeptide; tombusvirus pl9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pi 30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
  • the dsRNA is greater than 21 nucleotides in length. In some embodiments, the dsRNA is a hairpin RNA. In some embodiments, the bacterial cell expresses an RNase III polypeptide. In some embodiments, the bacterial cell expresses an RNase III polypeptide encoded by an exogenous nucleic acid sequence. In some embodiments, the bacterial cell is an Escherichia coli cell. In some embodiments, at least one of the siRNA-binding polypeptide and the dsRNA are constitutively expressed. In some embodiments, at least one of the siRNA-binding polypeptide and the dsRNA are inducibly expressed. In some embodiments, the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA is part of a plasmid.
  • a method of producing one or more siRNA species which can inhibit the expression of a target RNA comprising: culturing a bacterial cell as described herein under conditions suitable for the production of siRNAs.
  • the method further comprises a second step of isolating the siRNA-binding polypeptide and eluting the siRNAs bound to the siRNA-binding polypeptide.
  • the method further comprises purifying the siRNAs eluted from the siRNA-binding polypeptide by chromatography e.g. anion exchange HPLC.
  • the method further comprises contacting the cell with one or more modified nucleotides before or during the culturing step.
  • composition comprising a siRNA produced according to the methods described herein.
  • composition further comprises a population of siRNA species.
  • composition comprising a siRNA isolated from a bacterial cell as described herein.
  • the composition further comprises a population of siRNA species.
  • siRNA produced according to the methods described herein in the production of a medicament.
  • siRNA isolated from a bacterial cell of as described herein in the production of a medicament.
  • a vector comprising; a nucleic acid encoding a siRNA- binding polypeptide; and a dsRNA cloning site.
  • the dsRNA cloning site comprises at least one restriction enzyme site and can accept the insertion of at least one nucleic acid sequence such that a dsRNA is encoded and can be expressed.
  • a vector comprising: a nucleic acid encoding a siRNA-binding polypeptide; and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • the siRNA-binding polypeptide is selected from the group consisting of: pi 9 polypeptide; tombusvirus pl9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
  • the vector is a plasmid. In some embodiments, the plasmid further comprises a bacterial origin of replication.
  • a library of siRNA species comprising: a plurality of clonal bacterial cell populations; wherein each clonal population is comprises bacterial cells as described herein.
  • a library of siRNA species the library comprising: a plurality of populations of siRNAs; wherein each population of siRNAs is obtained according to the methods described herein.
  • each population of siRNAs binds to a single target RNA.
  • kits comprising a bacterial cell as described herein.
  • a kit for the production of one or more species of siRNA the kit comprising: a bacterial cell comprising an siRNA-binding polypeptide; and at least one vector comprising a dsRNA cloning site.
  • a kit for the production of one or more species of siRNA the kit comprising: a bacterial cell comprising an siRNA-binding
  • kits comprising a vector as described herein.
  • kit for the production of one or more species of siRNA the kit comprising two vectors; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and wherein the second vector comprises a dsRNA cloning site.
  • kits for the production of one or more species of siRNA comprising two plasmids; wherein the first vector comprises a nucleic acid encoding a siRNA- binding polypeptide; and wherein the second vector comprises a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • at least one vector is a plasmid.
  • the plasmid further comprises a bacterial origin of replication.
  • the kit further comprises a bacterial cell.
  • kits for the production of one or more species of siRNA comprising; a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA cloning site.
  • a kit for the production of one or more species of siRNA the kit comprising; a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • the siRNA-binding polypeptide comprises a purification tag.
  • the siRNA-binding polypeptide is encoded by a nucleic acid.
  • the bacterial cell expresses an RNase III polypeptide.
  • the cell is an Escherichia coli cell.
  • at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to a constitutive promoter.
  • at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to an inducible promoter.
  • the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA is part of a plasmid.
  • described herein is a kit comprising the library as described herein. BRIEF DESCRIPTION OF THE DRAWINGS
  • Figures 1A-1E demonstrate that ectopic pl9 expression captures small RNAs in E. coli.
  • Figure 1A depicts an image of a gel from experiments in which pl9-coupled magnetic beads were used to isolate small dsRNAs from total RNA isolated from mammalian ACH2 cells, or from E. coli cells, or from E. coli cells containing a pcDNA3.1-P19 expression plasmid. Captured RNAs were 5' 32 P-labeled, separated on a native polyacrylamide gel and detected by autoradiography.
  • Figure IB depicts images of gels from experiments in which expression of FLAG-tagged pi 9, but not TREX1 or empty plasmid (V, vector), from pcDNA3.1+ in E.
  • FIG. 1C depicts an image of a gel from experiments in which total RNAs purified from E. coli containing an empty vector, or pcDNA3.1+ expressing His- tagged pi 9 or His-tagged pl9 mutant proteins defective in RNA binding (Mutl 14 : W39G, W42G and Mut2 16 : K71A, R72G) were separated on a denaturing polyacrylamide gel and stained with SYBR Gold.
  • Figure ID depicts images of gels from experiments in which pl9-associated RNAs were isolated with pl9-coupled magnetic beads from total RNA extracted from WT E. coli (DH5a or MG1655 Alac) or RNase III mutant strains (in MG1655 Alac background) expressing the His-tagged pi 9 protein.
  • pl9-captured RNAs were separated on native or denaturing gels and stained with SYBR Gold, pi 9 expression was verified by His immunoblot.
  • the asterisk (*) indicates equal loading of a background band.
  • Figure IE depicts an image of a gel from experiments in which pl9-associated RNAs were isolated with pl9-coupled magnetic beads from total RNA extracted from pl9 expressing E.
  • Figures 2A-2F demonstrate pro-siRNAs knockdown EGFP expression.
  • Figure 2A depicts a schematic of pGEX-4T-l -pl9-T7 plasmid and the method to produce pro-siRNAs from E. coli.
  • Figure 2B depict an image of a gel from experiments in which anion exchange HPLC fractions of SDS-eluted RNAs (isolated from E. coli transformed to express pro-siRNAs) were separated on a native polyacrylamide gel and stained with SYBR Gold.
  • Figure 2C depicts images of gels from experiments in which nuclease sensitivity assay confirms pro-siRNAs are dsRNAs.
  • FIG. 2D depicts images of gels from experiments in which anti-Ago mAb 2A8 or mouse total IgG was used to immunoprecipitate RNAs in negative control (NC) siRNA or EGFPFL pro-siRNA-transfected HeLa-dlEGFP cells.
  • Immunoprecipitated RNAs were analyzed by Northern blot using a probe complementary to the full length EGFP coding sequence (top) or 5' 32 P end-labeling (middle). Bottom immunoblot is probed for Ago protein.
  • Figure 2E depicts a graph of the results of qRT-PCR of EGFP expression and EGFP mean fluorescence intensity by flow cytometry in HeLa-dlEGFP cells transfected with either 4 nM of siRNA or pro- siRNAs. Data are normalized to cells treated with negative control (NC) siRNA and are mean ⁇ SD of 2 (qRT-PCR) and 3 (EGFP fluorescence) independent experiments. mRNA level is relative to GADPH.
  • Figure 2F depicts images og gels from experiments in which HPLC-purified pro-siRNAs were separated on native or denaturing polyacrylamide gels stained with SYBR Gold.
  • Figures 3A-3D demonstrate that pro-siRNA-mediated knockdown of endogenous and viral gene expression in human cells.
  • Figure 3A depicts graphs and images of gels from experiments in which qRT-PCR and immunoblot assays of genes targeted for knockdown with the indicated siRNAs or pro-siRNAs, which were transfected (4 nM) into HeLa-dlEGFP (top) or HCT116 (bottom) cells.
  • Figure 3B depicts graphs of cell counts after transfection with PLK1 siRNA or pro-siRNA (4 nM) or negative control (NC) siRNA or EGFP pro-siRNA as nontargeting controls, respectively.
  • Figure 3C depicts a schematic and graphs of the results of experiments demonstrating inhibition of HIV-1 spreading by transfection of vif siRNAs and pro-siRNAs (4 nM).
  • vif siRNAs were either individually transfected (vif siRNA- 1 and vif siRNA-2) or co-transfected with 2 nM each of vif siRNA- 1 and siRNA-2 (vif siRNAl+2).
  • Figure 3D depicts graphs of the suppression of multiple HIV-1 strains by gag pro-siRNAs (transfected at 20 nM).
  • gagB siRNA from IIIB and its corresponding sequences in UG29 and ⁇ 22 strains were shown, (left) bar graph is gag mRNA knockdown in HeLa-CD4 cells (for IIIB), U87.CD4.CXCR4 cells (for UG29) and U87.CD4.CCR5 cells (for IN22); (right) infectivity of culture supernatants by TZM-bl assay. Data are mean ⁇ SD of 3 ( Figures 3A-3C) and 2 ( Figure 3D) independent experiments. mRNA expression and TZM-bl luciferase data are normalized to cells transfected with NC siRNA. Figure 3D discloses SEQ ID NOS 123-125, respectively, in order of appearance.
  • Figures 4A-4F demonstrating pro-siRNA sequences and tests of off-target effects.
  • Figure 4A depicts a graph of length distribution of EGFPFL, EGFP 100 and LMNA pro-siRNAs assessed by deep sequencing.
  • Figure 4B depicts a graph of the percentage of sequence content of all aligned deep sequencing reads.
  • Figure 4C depicts graphs of the distribution of aligned deep sequencing reads of EGFPFL, EGFP 100 and LMNA pro-siRNAs.
  • Figure 4D depicts volcano plots of expression changes versus p value of all annotated transcripts detected by RNA deep sequencing in HeLa-dlEGFP cells transfected with EGFP siRNAs or pro-siRNAs relative to expression in cells transfected with a negative control (NC) siRNA. Arrows indicate EGFP and the number is its fold change. Cut-off of significance is q_value ⁇ 0.05 (default in Cufflinks).
  • Figure 4E depicts volcano plots of expression changes (1.2 fold less or more) versus p value detected by microarray in HeLa-dlEGFP cells transfected with LMNA siRNAs or pro-siRNAs relative to expression in cells transfected with a NC siRNA. Arrows indicate LMNA and the number is its fold change. Cut-off of significance is p ⁇ Q.Q5 (by paired T-test).
  • Figure 4F depicts a graph of the percentage of significantly changed transcripts in Figures 4D-4E.
  • Figures 5A-5B demonstrate that ectopic expression of pi 9 stabilizes -21 nt small RNA species in Listeria monocytogenes.
  • Figure 5A depicts an image of gel demonstrating that approximately 21 nt small RNAs co-purify with pi 9.
  • L. monocytogenes was transformed with an empty vector (pLIV-1) or with pLIV-1 encoding inducible N-terminal His-tagged pi 9 (pLIV-l-pl9- His).
  • Duplicate cultures were grown in the presence of IPTG to induce protein expression. Samples were then lysed and incubated with Ni resin to purify the His-pl9 protein and any associated RNAs.
  • RNA markers RNA markers. Arrow indicates -21 nt small RNAs.
  • Figure 5B depicts an image of an immunoblot with His antibody to confirm IPTG-dependent induction of His-pl9.
  • Figures 6A-6D demonstrate that SDS efficiently elutes GST-pl9-His-bound small RNAs but not GST-pl9-His protein.
  • Figure 6A depicts an image of gel from an experiment in which GST- pl9-His protein, induced in E. coli with IPTG and purified by imidazole elution from Ni resin, was assayed by SDS-PAGE and Coomassie blue staining.
  • Figure 6B depicts an image of a gel demonstrating that imidazole, but not SDS (0.5%), elutes GST-pl9-His protein from Ni beads. Coomassie blue staining of proteins eluted from Ni resin with imidazole (lane 1) or SDS (lane 2).
  • Lane 3 shows proteins bound to the Ni resin before any elution
  • lane 4 is a sample of the SDS eluate
  • lane 5 shows proteins remaining on the beads after SDS elution (lane 5).
  • M protein markers.
  • Figure 6C depicts an image of a gel demonstrating that SDS elution efficiently elutes GST-pl9-His- bound small RNAs.
  • Ni resin was boiled before or after incubation with 0.5% SDS and bound RNAs were analyzed on a denaturing polyacrylamide gel stained with SYBR Gold. Arrow indicates -21 nt small RNAs, which were removed by SDS treatment.
  • Figure 6D depicts a schematic summary of the effect of imidazole or SDS elution of material captured by Ni resin from E. coli expressing GST-pl9- His protein.
  • Figures 7A-7B demonstrate the dose response comparison of gene silencing by EGFP siRNAs and pro-siRNAs and test of antisense EGFP construct.
  • Figure 7A depicts a graph of EGFP fluorescence in HeLa-dlEGFP cells transfected with either siRNAs or pro-siRNAs at the indicated concentrations. Data are a representative dose-response experiment.
  • Figure 7B depicts schematics and a graph. Schemes of empty, EGFP antisense and hairpin plasmids used to produce pro-siRNAs. Bar graph is the percentage of EGFP expressing HeLa-dlEGFP cells after transfection of NC siRNA and pro-siRNAs (at 0.5 tiM). Data are mean ⁇ SD of 2 independent transfections.
  • Figures 8A-8B demonstrate that pro-siRNA knockdown of gene expression is independent of Dicer.
  • Figure 8A depicts a graph of Negative control (NC) siRNA, EGFP siRNA or EGFPFL pro-siRNA co-transfected with pEGFP-Nl plasmid into HCT116 cells that contained a Dicer exon 5 deletion mutation (HCT116 Dicer ' ) .
  • Figure 8B depicts an image of a gel from experiments in which double stranded RNAs, siRNAs and pro-siRNAs were incubated or not with recombinant Dicer protein for 18 hrs at 37 °C. Resulting products were separated on a 20% polyacrylamide gel and stained with SYBR Gold.
  • Figure 9 depicts graphs demonstrating dose response comparison of gene silencing by pro-siRNAs and commercial siRNAs.
  • Total RNAs were extracted from HeLa-dlEGFP cells 24 hrs after transfection. mRNA levels were normalized to negative control siRNA transfected cells. Two independent experiments were shown.
  • Damachon siRNAs siRNA-Dl-D4.
  • siRNA of published sequence siRNA-G.
  • Figures 10A-10B demonstrate that pro-siRNAs induce little expression of proinflammatory cytokines in primary monocyte-derived human macrophages.
  • Figure 10A depicts a graph of the results of a qRT-PCR assay of the indicated proinflammatory cytokine gene mRNAs, 4 hr after treatment with the indicated concentrations of LPS, synthetic siRNAs, HPLC-purified pro- siRNAs or SDS-eluate. mRNA levels were normalized to levels in untreated cells.
  • Figure 10B depicts a graph of the results of a qRT-PCR assay of the indicated proinflammatory cytokine gene, LMNA and IFIT1 mRNAs, 24 hrs after transfection with indicated siRNA and pro-siRNA (at 20 nM). PolyLC was used as positive control and mRNA levels were normalized to levels in mock transfected cells.
  • Figure 11 depicts length profile and distribution of deep sequencing reads aligned to the pro-siRNA target sequences.
  • Figures 12A-12E demonstrate a test of strand bias and validation of pro-siRNA 'hot spots' for EGFPFL pro-siRNA.
  • Figure 12A depicts a graph of the position of DNA oligonucleotides (26-27 nt) used for probing EGFPFL small RNAs compared to position of aligned sequencing reads. The linear scale emphasizes sequencing hot spots.
  • F forward probe: R, reverse probe.
  • Figure 12B depicts images of gels from experiments in which purified pro-siRNAs were denatured and incubated with the indicated DNA probes, and then the reaction mixture was analysed for the formation of DNA:RNA hybrids by native polyacrylamide gel electrophoresis and autoradiography, (top) short exposure; (middle) long exposure; (bottom) DNA oligonucleotides only, exposed for 1 hr, to show comparable labelling. Arrows indicate the DNA:RNA hybrids.
  • Figure 12C depicts a graph of band intensities from Figure 12B which were quantified using Multi-gauge software (Fujifilm);
  • Figure 12D depicts a graph of the ratio of sense to antisense signal for each pair of probes calculated by dividing the DNA:RNA hybrid band intensities detected with the "R" oligonucleotide by that detected with the "F” oligonucleotide.
  • Figure 12E depicts a graph of normalized levels (to Sil) of hybridization signals (from Figure 12C) and numbers of deep sequencing reads (from Table 2) of the three hot spots.
  • Figures 13A-13E demonstrate the similarity of EGFPFL pro-siRNA sequence contents and hot spot patterns obtained in two independent pro-siRNA preparations. Graphs are depicted, comparing gene knockdown of EGFP measured by flow cytometry (Figure 13 A) sequence content ( Figure 13B), length profile ( Figure 13C) and distribution ( Figure 13D) of deep sequencing reads of two independent EGFPFL pro-siRNAs ⁇ EGFPFL-1 and EGFPFL-2).
  • Figure 13E depicts a graph comparing deep sequencing reads profiles of pro-siRNAs made from top (1-360 nt, Hotspot-1) or bottom half of EGFP (361-720 nt, Hotspot-2) with pro-siRNAs made from full length EGFP (1-720 nt, EGFPFL-1).
  • NC negative control siRNA
  • Figures 14A-14D demonstrate the off-target effect of siRNAs and pro-siRNAs.
  • Figure 14D depicts Venn diagrams for significantly changed genes in HeLa-dlEGFP cells transfected with EGFP siRNAs or pro-siRNAs.
  • Figure 14B depicts volcano plots of expression changes versus p value of all annotated lincRNA by RNA deep sequencing in HeLa-dlEGFP cells transfected with EGFP siRNAs or pro-siRNAs relative to expression in cells transfected with a negative control (NC) siRNA. Cut-off of significance is q_value ⁇ 0.05 (default in Cufflinks).
  • Figure 14C depicts graphs of the number of significantly changed lincRNAs.
  • Figure 14D depicts Venn diagrams for significantly changed genes in HeLa-dlEGFP cells transfected with LMAN siRNAs or pro-siRNAs.
  • Figures 15A-15C demonstrate a two-plasmid alternate method for generating pro-siRNAs in E. coli.
  • Figure 15A depicts a schematic of method to produce pro-siRNAs in E. coli using a two plasmid approach, where one plasmid directs pi 9 expression and the other expression of dsRNA corresponding to the target sequence.
  • Figure 15B depicts and image of a gel from experiments in which SDS eluate of pro-siRNAs targeting EGFP, produced using this two-plasmid approach from bacteria transformed with either pRSF-GST-pl9-His or pCDF-GST-pl9-His (encoding GST-pl9-His fusion protein) in combination with L4440-EGFP plasmid (encoding T7-driven sense and antisense EGFP transcripts), were separated on a native polyacrylamide gel stained with SYBR Gold.
  • Figure 15C depicts graphs of EGFP fluorescence measured by flow cytometry in HeLa-dlEGFP cells transfected with indicated siRNA or pro-siRNA (-10 nM). Data are representative of 3 independent experiments.
  • Figure 16 demonstrates an exemplary method to improve yield of pro-siRNAs.
  • pGEX- 4T-l-pl9-T7 plasmid containing EGFP hairpin (used to make EGFPFL pro-siRNA) was co- transfected with pi 9 overexpressing plasmids (pCDF-pl9 or pRSF-pl9) or E. coli RNase III overexpressing plasmid (pCDF-RNase III).
  • the two-plasmid system ( Figures 15A-15C) of co- transfecting L4440-EGFP with pCDF-pl9 or pRSF-pl9 was also tested. All E. coli cells were cultured under the same conditions.
  • pro-siRNAs were produced as in Figure 2A and equal proportions of SDS eluate were separated on a native polyacrylamide gel and stained with SYBR Gold. -21 nt small RNA band was quantified using Gel Logic software and signals were normalized to the band in the first sample lane.
  • Embodiments of the invention described herein are directed to methods and compositions relating to the production of siRNAs in vivo, e.g. in bacterial cells (siRNAs produced according to the methods and compositions described herein are also referred to herein as "pro-siRNAs").
  • the technology described herein is derived from the inventors' discovery that prokaryotic cells have the ability to generate siRNAs (e.g. pro-siRNAs).
  • pro-siRNAs As prokaryotic cells are not known to express components of the canonical RNAi machinery (e.g. Dicer), it was previously believed that prokaryotic cells were incapable of producing siRNAs.
  • pi 9 polypeptide As described in the Examples herein, when the inventors isolated pi 9 polypeptide which was expressed in a prokaryotic cell, it was found that the pi 9 polypeptide was bound to siRNAs present in the prokaryotic cell (pro-siRNAs). In the absence of the exogenous pi 9 polypeptide, these siRNAs are undetectable. These results indicated, in contrast to existing consensus in the field, that prokaryotic cells are capable of generating siRNAs, even in the absence of the canonical siRNA machinery, e.g. Dicer. The results further indicate that the endogenous siRNAs have exceptionally short half-lives which prevent their detection and/or isolation.
  • siRNAs specific for the target RNA were generated by the prokaryotic cell.
  • the activity of these siRNAs in silencing the target RNA expressed by a eukaryotic cell is demonstrated herein.
  • Embodiments described herein use endogenous biological processes to generate siRNAs from dsRNA, not requiring the use of algorithms which attempt to predict efficacious siRNA sequences.
  • Embodiments described herein also relate to populations of multiple siRNA species, wherein the population as a whole is specific for a target RNA. Such populations of siRNA species can have reduced off-target effects and greater efficacy than single RNA species.
  • the terms “decrease”, “reduced”, “reduction”, or “inhibit” are all used herein to mean a decrease by a statistically significant amount.
  • the terms “reduced”, “reduction”, “decrease”, or “inhibit” can mean a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or more or any decrease of at least 10% as compared to a reference level.
  • the terms can represent a 100%> decrease, i.e.
  • a non-detectable level as compared to a reference level.
  • a marker or symptom is meant a statistically significant decrease in such level.
  • the decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is preferably down to a level accepted as within the range of normal for an individual without such disorder.
  • the terms “increased”, “increase”, “enhance”, or “activate” are all used herein to mean an increase by a statically significant amount.
  • the terms “increased”, “increase”, “enhance”, or “activate” can mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%), or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100%) increase or any increase between 10-100%) as compared to a reference level, or at least about a 2-fold, or at least about a 3 -fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • a "subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters.
  • Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon.
  • the subject is a mammal, e.g., a primate, e.g., a human.
  • the terms, "individual,” “patient” and “subject” are used interchangeably herein.
  • the subject is a mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but is not limited to these examples.
  • a subject can be male or female.
  • a "subject in need" of treatment for a particular condition can be a subject having that condition, diagnosed as having that condition, or at risk of developing that condition.
  • proteins and “polypeptides” are used interchangeably herein to designate a series of amino acid residues connected to the other by peptide bonds between the alpha- amino and carboxy groups of adjacent residues.
  • the terms “protein”, and “polypeptide” refer to a polymer of amino acids, including modified amino acids (e.g., phosphorylated, glycated, glycosylated, etc.) and amino acid analogs, regardless of its size or function.
  • modified amino acids e.g., phosphorylated, glycated, glycosylated, etc.
  • polypeptide are often used in reference to relatively large polypeptides, whereas the term “peptide” is often used in reference to small polypeptides, but usage of these terms in the art overlaps.
  • protein and “polypeptide” are used interchangeably herein when referring to a gene product and fragments thereof.
  • exemplary polypeptides or proteins include gene products, naturally occurring proteins, homologs, orthologs, paralogs, fragments and other equivalents, variants, fragments, and analogs of the foregoing.
  • nucleic acid or “nucleic acid sequence” refers to any molecule, preferably a polymeric molecule, incorporating units of ribonucleic acid, deoxyribonucleic acid or an analog thereof.
  • the nucleic acid can be either single-stranded or double-stranded.
  • a single-stranded nucleic acid can be one strand nucleic acid of a denatured double- stranded DNA. Alternatively, it can be a single-stranded nucleic acid not derived from any double-stranded DNA.
  • the template nucleic acid is DNA.
  • the template is RNA.
  • Suitable nucleic acid molecules are DNA, including genomic DNA or cDNA. Other suitable nucleic acid molecules are RNA, including mRNA.
  • RNA transcribed from a gene and polypeptides obtained by translation of mRNA transcribed from a gene.
  • gene means the nucleic acid sequence which is transcribed (DNA) to RNA in vitro or in vivo when operably linked to appropriate regulatory sequences.
  • a gene may or may not include regions preceding and following the coding region, e.g. 5' untranslated (5'UTR) or "leader” sequences and 3' UTR or “trailer” sequences, as well as intervening sequences (introns) between individual coding segments (exons).
  • the term "operatively linked” includes having an appropriate start signal (e.g., ATG) in front of the polynucleotide sequence to be expressed, and maintaining the correct reading frame to permit expression of the polynucleotide sequence under the control of the expression control sequence, and, optionally, production of the desired polypeptide encoded by the polynucleotide sequence.
  • transcription of a nucleic acid modulatory compound is under the control of a promoter sequence (or other transcriptional regulatory sequence) which controls the expression of the nucleic acid in a cell- type in which expression is intended. It will also be understood that the modulatory nucleic acid can be under the control of transcriptional regulatory sequences which are the same or which are different from those sequences which control transcription of the naturally-occurring form of a protein.
  • isolated refers, in the case of a nucleic acid or polypeptide, to a nucleic acid or polypeptide separated from at least one other component ⁇ e.g. , nucleic acid or polypeptide) that is present with the nucleic acid or polypeptide as found in its natural source and/or that would be present with the nucleic acid or polypeptide when expressed by a cell, or secreted in the case of secreted polypeptides.
  • a chemically synthesized nucleic acid or polypeptide or one synthesized using in vitro transcription/translation is considered “isolated.”
  • exogenous refers to a substance (e.g. a nucleic acid or polypeptide) present in a cell other than its native source.
  • exogenous can refer to a nucleic acid or a protein (that has been introduced by a process involving the hand of man into a biological system such as a cell or organism in which it is not normally found or in which it is found in undetectable amounts.
  • a substance can be considered exogenous if it is introduced into a cell or an ancestor of the cell that inherits the substance.
  • endogenous refers to a substance that is native to the biological system or cell.
  • complementary or “complementary base pair” refers to A:T and G:C in DNA and A:U in RNA.
  • DNA consists of sequences of nucleotide only four nitrogenous bases: base or base adenine (A), thymine (T), guanine (G), and cytosine (C). Together these bases form the genetic alphabet, and long ordered sequences of them contain, in coded form, much of the information present in genes.
  • Most RNA also consists of sequences of only four bases. However, in RNA, thymine is replaced by uridine (U).
  • substantially complementary refers to a first nucleotide sequence having at least 90% complementarity over the entire length of the sequence with a second nucleotide sequence, e.g. 90%> complementary, 95% complementary, 98% complementary, 99% complementary, or 100%) complementary.
  • Two nucleotide sequences can be substantially complementary even if less than 100%) of the bases are complementary, e.g. the sequences can be mismatched at certain bases.
  • RNAi gene silencing
  • RNAi agent can be substantially homologous to the target RNA gene or genomic sequence, or a fragment thereof.
  • dsRNA double-stranded RNA
  • RNAs suitable for inhibiting or interfering with the expression of a target RNA include RNA derivatives and analogs.
  • RNAi can be caused by any type of interfering RNA, including but are not limited to, siRNA, shRNA, endogenous microRNA and artificial microRNA.
  • the RNAi molecule is a small interfering RNA (siRNA).
  • An RNAi agent can cause a decrease in the level of a target RNA in a cell by at least about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%) or more of the target RNA level found in the cell without the presence of the gene silencing agent.
  • the target RNA levels are decreased by at least about 70%, about 80%, about 90%, about 95%, about 99% or more.
  • the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder.
  • treating includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder.
  • Treatment is generally “effective” if one or more symptoms or clinical markers are reduced.
  • treatment is “effective” if the progression of a disease is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
  • the term "pharmaceutical composition” refers to the active agent in combination with a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • pharmaceutically acceptable is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • administering refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site.
  • Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
  • compositions, methods, and respective component(s) thereof are used in reference to compositions, methods, and respective component(s) thereof, that are essential to the method or composition, yet open to the inclusion of unspecified elements, whether essential or not.
  • Consisting essentially of refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment.
  • the present invention can be performed using standard procedures, as described, for example in Sambrook et al., Molecular Cloning: A Laboratory Manual (3 ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (2001); Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (1995); Current Protocols in Cell Biology (CPCB) (Juan S. Bonifacino et. al. ed., John Wiley and Sons, Inc.), and Culture of Animal Cells: A Manual of Basic Technique by R. Ian Freshney, Publisher: Wiley- Liss; 5th edition (2005), Animal Cell Culture Methods (Methods in Cell Biology, Vol. 57, Jennie P. Mather and David Barnes editors, Academic Press, 1st edition, 1998) which are all incorporated by reference herein in their entireties.
  • Embodiments of the technology described herein include methods and compositions relating to a bacterial cell comprising a siRNA-binding polypeptide and a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially complementary to at least one target RNA.
  • siRNA-generating enzymes e.g. RNAses
  • present in the bacterial cell e.g. either endogenous or exogenous
  • siRNA-binding polypeptide can enable purification of the siRNA molecules from the other constituents of the bacterial cell and prevent further degradation of the siRNA to non-siRNA substituents, e.g. dsRNAs of less than 15 nucleotides in length or individual ribonucleotides.
  • double stranded RNA or “dsRNA” refers to RNA molecules that comprise two substantially complementary strands. Double-stranded molecules include those comprising a single RNA molecule that doubles back on itself to form a two-stranded structure, e.g. a stem-loop molecule or a hairpin molecule.
  • a dsRNA can comprise nucleic acid sequences which are not substantially complementary to other sequences of the dsRNA (i.e. a single- stranded portion of the dsRNA), for example, the loop part of a stem-loop structure.
  • the portion of the dsRNA which comprises a nucleic acid sequence substantially complementary to a target RNA should comprise, at least in part, the double-stranded portion of a dsRNA.
  • the double-stranded portion of a dsRNA comprising a nucleic acid sequence substantially complementary to at least one target RNA can be 21 nucleotides in length or greater, e.g.
  • 21 nucleotides or greater 22 nucleotides or greater, 23 nucleotides or greater, 24 nucleotides or greater, 25 nucleotides or greater, 50 nucleotides or greater, 100 nucleotides or greater, 200 nucleotides or greater, 500 nucleotides or greater, 1000 nucleotides or greater in length.
  • target RNA refers to a RNA present in a cell (i.e. the "target cell”).
  • the target RNA comprises a target sequence to which one strand of a siRNA according to the methods and compositions described herein binds, thereby causing RNAi silencing of the target RNA.
  • the target cell can be the bacterial cell comprising a siRNA-binding polypeptide or another cell, either prokaryotic or eukaryotic.
  • the target sequence can be an RNA that can be translated (i.e. it can encode a polypeptide, e.g. mRNA) or it can be an RNA that is not translated (i.e. a non-coding RNA).
  • the target sequence can be any portion of an mRNA. In some embodiments, the target sequence can be a sequence endogenous to the cell. In some embodiments, the target sequence can be a sequence exogenous to the cell. In some embodiments, the target sequence can be sequence from an organism that is pathogenic to the target cell, e.g. the target sequence can be sequence from a viral, bacterial, fungal, and/or parasitic origin. In some embodiments, the target sequence is a viral nucleotide sequence.
  • a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence substantially complementary to part or all of the sequence of the target RNA.
  • a dsRNA comprising a nucleic acid sequence
  • substantially complementary to a target RNA can comprise sequence complementary to part or all of a specific allele, variant, and/or mutation (e.g., insertions, deletions, fusions, SNPs, etc.) of a target RNA.
  • the dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise nucleic acid sequence(s) substantially complementary to multiple target RNAs (e.g. target RNAs encoding separate genes or target RNAs encoding multiple variants of the same gene).
  • a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence substantially complementary to all or part of one or more exons of a target mRNA.
  • a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence substantially complementary to a cDNA.
  • a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence (or its complement) obtained from the transcriptome and/or genome of a cell.
  • the dsRNA can comprise two separate complementary strands, e.g. a sense and antisense strand.
  • the dsRNA can be a hairpin RNA, i.e. an RNA comprising two portions which are reverse complements, separated by a sequence which will not self-anneal, thus forming a stem-loop or "hairpin" structure.
  • the double-stranded portion of a hairpin RNA can be at least 19 nucleotides in length.
  • the double-stranded portion of a hairpin RNA can be at least 25 nucleotides in length.
  • the double- stranded portion of a hairpin RNA can be 30 nucleotides in length or greater, e.g.
  • the dsRNA can be a shRNA.
  • shRNA small hairpin RNA
  • these shRNAs are composed of a short, e.g. about 19 to about 25 nucleotide, antisense strand, followed by a nucleotide loop of about 5 to about 9 nucleotides, and the analogous sense strand.
  • the sense strand can precede the nucleotide loop structure and the antisense strand can follow.
  • one strand of the double-stranded portion of a dsRNA can correlate with a decreased level of off-target effects, e.g. silencing of non-targeted genes.
  • one strand of the double-stranded portion of a dsRNA can be at least 100 nucleotides in length.
  • one strand of the double-stranded portion of a dsRNA can be at least 100 nucleotides in length, at least 200 nucleotides in length, at least 300 nucleotides in length, at least 400 nucleotides in length, at least 500 nucleotides in length, at least 700 nucleotides in length, or at least 1000 nucleotides in length.
  • the dsRNA can be exogenous to the cell.
  • the target sequence of the target RNA can be exogenous to the cell.
  • the target RNA can be exogenous to the cell.
  • the nucleic acid sequence substantially complementary to a target RNA can be exogenous to the cell.
  • siRNAs can be generated from the dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • siRNA refers to a nucleic acid that forms an RNA molecule comprising two individual strands of RNA which are substantially complementary to each other.
  • the siRNA is at least about 15-40 nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is about 15-40 nucleotides in length, and the double stranded siRNA is about 15-40 base pairs in length, preferably about 19-25 base nucleotides, e.g., 19, 20, 21, 22, 23, 24, or 25 nucleotides in length).
  • a siRNA can be blunt-ended.
  • a siRNA can comprise a 3' and/or 5' overhang on each strand having a length of about 0, 1, 2, 3, 4, or 5 nucleotides. The length of the overhang is independent between the two strands, i.e., the length of the overhang on one strand is not dependent on the length of the overhang on the second strand.
  • the siRNA molecules can also comprise a 3 ' hydroxyl group.
  • the siRNA can comprise a 5' phosphate group.
  • a siRNA has the ability to reduce or inhibit expression of a gene or target RNA when the siRNA is present or expressed in the same cell as the target gene, e.g. the target RNA.
  • siRNA-dependent post-transcriptional silencing of gene expression involves cutting the target RNA molecule at a site guided by the siRNA.
  • a single siRNA species can be generated from a dsRNA.
  • multiple siRNA species can be generated from a dsRNA.
  • two or more siRNA species can be generated from a dsRNA, e.g. two or more siRNA species, three or more siRNA species, five or more siRNA species, or ten or more siRNA species.
  • the term "a siRNA species" refers to one or more siRNA molecules which are identical in sequence.
  • the species can comprise sequence complementary to the same target RNA or to separate target RNAs.
  • a single dsRNA can comprise sequence complementary to multiple target RNAs.
  • a single dsRNA can comprise multiple sequences, each of which is
  • RNA complementary to a unique target RNA, e.g. a multiplicity of siRNA species targeting (e.g.
  • RNA complementary to a multiplicity of target RNAs can be generated from a single dsRNA.
  • a dsRNA present within a bacterial cell can be cleaved to generate one or more siRNA species.
  • the siRNA molecules can then be bound by a siRNA-binding polypeptide also present within the bacterial cell.
  • siRNA-binding polypeptide refers to a polypeptide capable of binding to siRNAs and increasing the half-life or detectable level of siRNAs in a prokaryotic cell.
  • the siRNA- binding polypeptide can bind preferentially or specifically to siRNAs as compared to other dsRNA species, e.g.
  • the polypeptide can bind preferentially or specifically to siRNAs as compared to dsRNAs greater than 25 or less than 15 nucleotides in size. In some embodiments, the siRNA-binding polypeptide can bind preferentially or specifically to siRNAs as compared to other dsRNA species, e.g. dsRNAs greater than 25 or less than 15 nucleotides in size. In one embodiment, the siRNA- binding polypeptide does not bind to dsRNA having a double-stranded portion longer than 25 nucleotides in length. In some embodiments, the siRNA-binding polypeptide can bind preferentially or specifically to siRNAs as compared to single-stranded RNA species.
  • a siRNA-binding polypeptide can detectably bind to a siRNA.
  • a siRNA-binding polypeptide can be a polypeptide that when expressed in a bacterial cell, can cause detectable levels of siRNAs to be present in that cell when detectable levels of siRNAs are not present in the wild-type bacterial cell.
  • a siRNA-binding polypeptide can be a polypeptide that increases the half-life or detectable level of siRNAs in a prokaryotic cell by at least 5%, e.g. by at least 5%, by at least 10%, by at least 20%, by at least 30%, by at least 50%, by at least 75%, by at least 100%, by at least 200% or more.
  • a siRNA-binding polypeptide can be a pi 9 polypeptide.
  • the term "pi 9" refers to a viral protein which binds specifically to dsRNAs and which suppresses RNAi-mediated host plant viral defenses.
  • the sequences of pi 9 polypeptides from a number of species are known, e.g. tombusvirus pl9 (NCBI Gene ID: 1493957; SEQ ID NO: l).
  • the pl9 polypeptide can be tombusvirus pl9.
  • pl9 homologues include Carnation Italian ringspot virus PI 9; Tomato bushy stunt virus pi 9; Artichoke mottled crinkle virus pl9; Lisianthus necrosis virus pl9; Pear latent virus pl9; Cucumber Bulgarian virus pl9; Cucumber necrosis virus pl9; Pelargonium necrotic spot virus pl9; Cymbidium ringspot virus pl9; Lisianthus necrosis virus pl9; Lettuce necrotic stunt virus pl9; Maize necrotic streak virus pl9; Grapevine Norwayn necrosis virus pl9; and Grapevine Francen latent virus pl9.
  • a pl9 polypeptide can comprise mutants, variants, homologues, and functional fragments of wildtype pi 9 polypeptides.
  • siRNA-binding polypeptide can include the Flock house virus B2; HC-Pro; Tobacco etch virus HC-Pro; P38; P122; P130; Tobamovirus P122/P130; p21 ; Rice hoja blanca tenuivirus (RHBV) NS3; Cucumber vein yellowing virus Plb; HC-Pro of potyviruses; p21 of Beet yellows virus and Closterovirus; and variants, homologues, or functional fragments of the foregoing.
  • an siRNA-binding polypeptide can be an enzymatically inactive member of the RISC complex, e.g. an enzymatically inactive variant or mutant of Argonaute or Dicer (see, e.g. Buker et al. Nat Struct Mol Bio 2007 14:200-7 and Liu et al. Molecular Cell 2012 46: 1-11 ; which are incorporated by reference herein in their entireties).
  • the siRNA- binding polypeptide is not an enzymatically active member of the RISC complex, e.g. an Argonaute or RISC polypeptide.
  • RISC complex refers to the proteins and single- stranded polynucleotides that interact to recognize target RNA molecules. Demonstrated components of RISC include the Argonaute proteins (e.g. Aubergine, Argonaute 2), R2D2, and Dicer (e.g. Dcr-2). In the case of an active RISC complex loaded with a single-stranded guide RNA derived from a siRNA, the RISC complex can cleave the target RNA molecule.
  • Argonaute proteins e.g. Aubergine, Argonaute 2
  • R2D2D2 Dicer
  • Dicer e.g. Dcr-2
  • the RISC complex can cleave the target RNA molecule.
  • a siRNA-binding polypeptide can be a polypeptide that can bind to nucleic acids, e.g. protamine, or a variant, homologue, or functional fragment thereof (see, e.g. Rossi. Nature Biotechnology 2005 23:682-4 and Reischl et al. Scientia Pharmaceutica 2010 78:686; which are incorporated by reference herein in their entirety).
  • a siRNA-binding polypeptide can be a polypeptide that can be bind to dsRNAs, e.g. TARBP2 or a polypeptide comprising a double-stranded RNA binding domain (see, e.g. US Patent Publication 2009/0093026; which is incorporated by reference herein in its entirety) or a variant, homologue, or functional fragment thereof.
  • a functional fragment of a siRNA-binding polypeptide can be any portion of a siRNA- binding polypeptide which retains at least 50% of the wild-type level of siRNA binding activity, e.g. at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or more.
  • a siRNA-binding polypeptide can comprise a purification tag.
  • purification tag refers to any peptide sequence suitable for purification of a siRNA-binding polypeptide, and optionally, siRNAs bound by the siRNA-binding polypeptide.
  • the purification tag specifically binds to (or is bound by) another moiety with affinity for the purification tag.
  • moieties which specifically bind to a purification tag can be attached to a matrix or a resin, e.g. agarose beads.
  • Moieties which specifically bind to purification tags can include antibodies, nickel or cobalt ions or resins, biotin, amylose, maltose, and cyclodextrin.
  • Exemplary purification tags can include histidine tags (such as a hexahistidine peptide (SEQ ID NO: 122)), which will bind to metal ions such as nickel or cobalt ions. Therefore, in certain embodiments the purification tag can comprise a peptide sequence which specifically binds metal ions.
  • Histidine tags such as a hexahistidine peptide (SEQ ID NO: 122)
  • the purification tag can comprise a peptide sequence which specifically binds metal ions.
  • Other exemplary purification tags are the myc tag (EQKLISEEDL (SEQ ID NO:3)), the Strep tag
  • purification tag also includes "epitope tags", i.e. peptide sequences which are specifically recognized by antibodies.
  • Exemplary epitope tags can include the FLAG tag, which is specifically recognized by a monoclonal anti-FLAG antibody.
  • the peptide sequence recognized by the anti-FLAG antibody consists of the sequence DYKDDDDK (SEQ ID NO: 5) or a substantially identical variant thereof. Therefore, in certain embodiments the purification tag can comprise a peptide sequence which is specifically recognized by an antibody.
  • purification tag also includes substantially identical variants of purification
  • siRNA- binding polypeptide can be a pi 9 fusion protein as described in US Patent Publication 2010/0209933; which is incorporated herein by reference in its entirety.
  • the siRNA-binding polypeptide can be encoded by a nucleic acid present in the bacterial cell, i.e. the polypeptide is transcribed and translated by the bacterial cell.
  • the siRNA-binding polypeptide can be introduced into the bacterial cell as a polypeptide. Uptake of polypeptides can be induced by any means in the art. Non-limiting examples include the protocols described in Shellman and Pettijohn. J Bacteriology 1991 173:3047-3059; which is incorporated by reference herein in its entirety.
  • a nucleic acid encoding a siRNA-binding polypeptide and/or a nucleic acid encoding a dsRNA can be present within the bacterial genome, e.g. the nucleic acids can be incorporated into the genome.
  • a nucleic acid encoding a siRNA-binding polypeptide and/or a nucleic acid encoding a dsRNA can be present within a vector.
  • a nucleic acid encoding a siRNA-binding polypeptide and/or a nucleic acid encoding a dsRNA can be present within portions of the same vector.
  • the nucleic acids encoding the siRNA-binding polypeptide and the dsRNA can be present within portions of separate vectors.
  • a vector refers to a nucleic acid construct designed for delivery to a host cell or transfer between different host cells.
  • a vector can be viral or non- viral. Many vectors useful for transferring exogenous genes into target cells are available, e.g. the vectors may be episomal, e.g., plasmids, virus derived vectors or may be integrated into the target cell genome, through homologous recombination or random integration.
  • a vector can be an expression vector.
  • expression vector refers to a vector that has the ability to incorporate and express heterologous nucleic acid fragments in a cell.
  • An expression vector may comprise additional elements, for example, the expression vector may have two replication systems, thus allowing it to be maintained in two organisms.
  • the nucleic acid incorporated into the vector can be operatively linked to an expression control sequence when the expression control sequence controls and regulates the transcription and translation of that polynucleotide sequence.
  • the dsRNA and the nucleic acid encoding the siRNA-binding polypeptide can be within the same operon. In some embodiments, the dsRNA and the nucleic acid encoding the siRNA-binding polypeptide can be within separate operons.
  • a siRNA-binding polypeptide and/or dsRNA encoded by a nucleic acid can be present within a portion of a plasmid.
  • Plasmid vectors can include, but are not limited to, pBR322, pBR325, pACYC177, pACYC184, pUC8, pUC9, pUC18, pUC19, pLG339, pR290, pKC37, pKClOl, SV 40, pBluescript II SK +/- or KS +/- (see "Stratagene Cloning Systems” Catalog (1993) from Stratagene, La Jolla, Calif, which is hereby incorporated by reference), pQE, pIH821, pGEX, pET series (see Studier et. al., "Use of T7 RNA Polymerase to Direct Expression of Cloned Genes," Gene Expression Technology, vol. 185 (1990), which is hereby
  • viral vector refers to a nucleic acid vector construct that includes at least one element of viral origin and has the capacity to be packaged into a viral vector particle.
  • the viral vector can contain a transgenic gene in place of non-essential viral genes.
  • the vector and/or particle may be utilized for the purpose of transferring any nucleic acids into cells either in vitro or in vivo.
  • Numerous viral vectors are known in the art and can be used as carriers of a nucleic acid into a cell, e.g. lambda vector system gtl 1, gt WES.tB, Charon 4.
  • siRNAs specific for the target RNA can be produced in a bacterial cell when both the dsRNA and the siRNA-binding polypeptide are present and/or expressed.
  • the dsRNA and/or the siRNA- binding polypeptide can be constitutively expressed.
  • nucleic acids encoding the dsRNA and/or the siRNA-binding polypeptide can be operably linked to a constitutive promoter.
  • the dsRNA and/or the siRNA-binding polypeptide can be inducibly expressed.
  • nucleic acids encoding the dsRNA and/or the siRNA-binding polypeptide can be operably linked to an inducible promoter.
  • an “inducible promoter” is one that is characterized by initiating or enhancing transcriptional activity when in the presence of, influenced by, or contacted by an inducer or inducing agent than when not in the presence of, under the influence of, or in contact with the inducer or inducing agent.
  • An “inducer” or “inducing agent” may be endogenous, or a normally exogenous compound or protein that is administered in such a way as to be active in inducing transcriptional activity from the inducible promoter.
  • the inducer or inducing agent e.g., a chemical, a compound or a protein
  • can itself be the result of transcription or expression of a nucleic acid sequence e.g., an inducer can be a transcriptional repressor protein
  • an inducer can be a transcriptional repressor protein
  • inducible promoters include but are not limited to, the lac operon promoter, a nitrogen-sensitive promoter, an IPTG- inducible promoter, a salt-inducible promoter, and tetracycline, steroid-responsive promoters, rapamycin responsive promoters and the like.
  • Inducible promoters for use in prokaryotic systems are well known in the art, see, e.g. the beta. -lactamase and lactose promoter systems (Chang et al., Nature, 275: 615 ( 1978, which is incorporated herein by reference); Goeddel et al.. Nature, 281 : 544 (1979), which is incorporated ereirs by reference), the arahinose promoter system, including the araBAD promoter (Guzman et al. J . Bacteriol., 174: 7716-7728 (1 992), which is incorporated herein by reference; Guzman et al., J.
  • An inducible promoter useful in the methods and systems as disclosed herein can be induced by one or more physiological conditions, such as changes in pH, temperature, radiation, osmotic pressure, saline gradients, cell surface binding, and the concentration of one or more extrinsic or intrinsic inducing agents.
  • the extrinsic inducer or inducing agent may comprise amino acids and amino acid analogs, saccharides and polysaccharides, nucleic acids, protein transcriptional activators and repressors, cytokines, toxins, petroleum-based compounds, metal containing compounds, salts, ions, enzyme substrate analogs, hormones, and combinations thereof.
  • the inducible promoter is activated or repressed in response to a change of an environmental condition, such as the change in concentration of a chemical, metal, temperature, radiation, nutrient or change in pH.
  • an inducible promoter useful in the methods and systems as disclosed herein can be a phage inducible promoter, nutrient inducible promoter, temperature inducible promoter, radiation inducible promoter, metal inducible promoter, hormone inducible promoter, steroid inducible promoter, and/or hybrids and combinations thereof.
  • Appropriate environmental inducers can include, but are not limited to, exposure to heat (i.e., thermal pulses or constant heat exposure), various steroidal compounds, divalent cations (including Cu2+ and Zn2+), galactose, tetracycline, IPTG (isopropyl- ⁇ - D thiogalactoside), as well as other naturally occurring and synthetic inducing agents and gratuitous inducers.
  • Inducible promoters useful in the methods and systems as disclosed herein also include those that are repressed by "transcriptional repressors” that are subject to inactivation by the action of environmental, external agents, or the product of another gene. Such inducible promoters may also be termed “repressible promoters” where it is required to distinguish between other types of promoters in a given module or component of the biological switch converters described herein. Preferred repressors for use in the present invention are sensitive to inactivation by physiologically benign agent.
  • a lac repressor protein is used to control the expression of a promoter sequence that has been engineered to contain a lacO operator sequence
  • treatment of the host cell with IPTG will cause the dissociation of the lac repressor from the engineered promoter containing a lacO operator sequence and allow transcription to occur.
  • a tet repressor is used to control the expression of a promoter sequence that has been engineered to contain a tetO operator sequence
  • treatment of the host cell with tetracycline will cause the dissociation of the tet repressor from the engineered promoter and allow transcription of the sequence downstream of the engineered promoter to occur.
  • a bacterial cell of the methods and compositions described herein can be any of any species.
  • the bacterial cells are of a species and/or strain which is amenable to culture and genetic manipulation.
  • the bacterial cell can be a gram-positive bacterial cell.
  • the bacterial cell can be a gram-negative bacterial cell.
  • the parental strain of the bacterial cell of the technology described herein can be a strain optimized for protein expression.
  • Non-limiting examples of bacterial species and strains suitable for use in the present technologies include Escherichia coli, E. coli BL21, E. coli Tuner, E. coli Rosetta, E. coli JM101, and derivatives of any of the foregoing.
  • a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can be processed to create siRNA molecules by a siRNA-generating enzyme (e.g. RNAse III) present within the bacterial cell.
  • a siRNA-generating enzyme e.g. RNAse III
  • the bacterial cell can be a cell which expresses a siRNA-generating polypeptide.
  • the bacterial cell can be a cell which overexpresses a siRNA-generating polypeptide.
  • siRNA-generating polypeptide refers to an enzyme with RNase activity which can cleave dsRNA in such a way that siRNAs result.
  • the siRNA-generating polypeptide can be an RNaselll polypeptide.
  • RNasel polypeptide refers to a eukaryotic class I RNase III, e.g. E. coli RNaselll (NCBI Gene ID: 947033; SEQ ID NO: 2).
  • siRNA-generating polypeptides can be mutants, variants, homologues, or functional fragments of wildtype siRNA-generating polypeptides which retain at least 50% of the siRNA generating activity of the wildtype, e.g.
  • the siRNA- generating enzyme can be endogenous to the bacterial cell. In some embodiments, the siRNA- generating enzyme can be exogenous to the bacterial cell.
  • a cell can comprise a mutation and/or transgene which enhances the expression and/or activity of a siRNA-generating polypeptide.
  • a cell can comprise a mutation in the endogenous RNaselll promoter which increases expression, or a cell can comprise a transgenic (e.g. exogenous) construct with an RNaselll gene under the control of a strong constitutive or inducible promoter, or a cell can comprise a nucleic acid encoding a polypeptide which increases the activity and/or expression of RNaselll, e.g. the T4 polynucleotide
  • a cell can express an ectopic level and/or amount of a siRNA-generating polypeptide (e.g. RNaselll).
  • siRNA-generating polypeptide e.g. RNaselll
  • ectopic refers to a substance that is found in an unusual location and/or amount. An ectopic substance can be one that is normally found in a given cell, but at a lower amount and/or at a different time.
  • the technology described herein relates to a method of producing one or more siRNA species which can inhibit the expression of a target RNA, the method comprising culturing a bacterial cell comprising at least a siRNA-binding polypeptide and a dsRNA wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA under conditions suitable for the production of siRNAs.
  • condition suitable for the production of siRNAs refers to conditions under which a siRNA-generating enzyme within a bacterial cell cleaves the dsRNA in the presence of a siRNA-binding polypeptide.
  • conditions suitable for the production of siRNAs can include conditions under which the cell will express (i.e. transcribe and, in some cases, translate) the dsRNA and/or the siRNA-binding polypeptide from the nucleic acid.
  • the precise conditions will vary depending on the exact identity of the bacterial cell, the presence of other exogenous DNA or mutations, and whether or not a nucleic acid encoding a dsRNA and/or siRNA-binding polypeptide is operably linked to an inducible or constitutive promoter.
  • conditions suitable for the production of siRNAs can include conditions which induce expression from the inducible promoter, e.g. permissive temperatures and/or the presence of compounds which induce expression from the inducible promoter.
  • conditions suitable for the production of siRNAs can include conditions which encourage exponential growth of the bacterial cells.
  • conditions suitable for the production of siRNAs in E. coli T7 Express Iq can include LB broth, Lennox (BD) at 37°C with shaking at 250 rpm and appropriate antibiotics.
  • a method of producing one or more siRNA species can further comprise isolating the siRNA-binding polypeptide and eluting the siRNAs bound to the siRNA- binding polypeptide.
  • the siRNA-binding polypeptide can be isolated via a purification tag as described elsewhere herein.
  • the siRNAs bound to a siRNA-binding polypeptide can be eluted from the isolated siRNA-binding polypeptide.
  • Methods of eluting nucleic acids from proteins are well known in the art.
  • siRNAs can be eluted from a siRNA-binding polypeptide by contacting the polypeptide-siRNA complex with a solution comprising 0.5% SDS for 10 min at room temperature with rotation. The solution can then be collected and passed through a 0.22 ⁇ centrifuge filter (Corning).
  • the siRNAs eluted from a siRNA-binding polypeptide can be further purified.
  • Methods of nucleic acid purification are well known in the art and include, but are not limited to anion exchange HPLC, PAGE purification, desalting, and filtration. See, e.g. Gjerde et al. "RNA Purification and Analysis” Wiley-VCH; 2009 and Farrell et al. "RNA Methodologies” 4 th Ed., Academic Press; 2010.
  • the siRNAs eluted from a siRNA-binding polypeptide can be further purified by HPLC.
  • the siRNAs can be isolated from the totality of the cell contents without first isolating the siRNAs bound to siRNA-binding polypeptides.
  • Methods of purifying RNA molecules are well known in the art, as described above, and any method or combination of methods known in the art can be used to isolate and/or purify the siRNAs produced according to the methods described herein.
  • the methods described herein can further comprise contacting the bacterial cell with one or more modified nucleotides before or during the culturing step, thereby causing one or more modified nucleotides to be incorporated into the siRNA(s) of the presently described technologies.
  • a modified nucleotide can be any nucleotide other than adenine "A”, guanine "G", uracil “U”, or cytosine "C”.
  • modified nucleotides include nucleotides which contains a modified sugar moiety, a modified phosphate moiety ami/or a modified nucleobase.
  • a modified nucleotide residue or a derivative or analog of a natural nucleotide are also useful.
  • modified residues, derivatives or analogues include, but are not limited to, aminoallyl UTP, pseudo- UTP, 5-I-UTP, 5-I-CTP, 5-Br-UTP, alpha-S ATP, alpha-S CTP, alpha-S GTP, alpha-S UTP, 4-thio UTP, 2-thio-CTP, 2TSTH2 UTP, 2TSTH2 CTP, and 2'F UTP.
  • modified nucleotides include, but are not limited to, aminoallyl uridine, pseudo-uridine, 5-I-uridine, 5-I-cytidine, 5-Br-uridine, alpha-S adenosine, alpha-S cytidine, alpha-S guanosine, alpha-S uridine, 4-thio uridine, 2-thio-cytidine, 2TSTH2 uridine, 2 ⁇ 2 cytidine, and 2' F uridine, including the free pho (NTP) RNA molecules as well as all other useful forms of the nucleotides.
  • NTP free pho
  • modified nucleotides can include ribonucleotides having a 2'-0-metbyl (TOMe), 2 i -deoxy-2 , fluoro, 2'-deoxy, 5-C-methyl, 2'-methoxyethyl, 4'-thio, 2'-amino, or 2'-C-allyl group, locked nucleic acid (LNA) nucleotides (e.g., LNA) nucleotides (e.g.
  • nucleotides having a nucleotide base analog such as. for example, C-phenyl, C- naphthyl, other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as
  • Modification of the sugar moiety can include, but is not limited to, replacement of the ribose ring with a hexose, cyelopentyl or cyclobexyl ring.
  • the D-ribose ring of a naturally-occurring nucleic acid can be replaced with an L- ribose ring or the (3-atiomer of a naturally occurring nucleic acid can be replaced with the a-anomer.
  • Modified phosphate moieties can include phosphorothioates, phosphorodithioates, methyl phosphorates, alkylphospbonates, alkylphosphonothioates, methyl phosphates, phosphoramidat.es, and the like, or combinations thereof.
  • Oligonucleotides which comprise such modified phosphate linkages can have impro ved properties when compared to corresponding oligonucleotides comprising only phosphate diester linkages, e.g. increased resistance to degradation by nucleases.
  • Modified nucieobases include 7-deazaguanine, 7-deaza-8-azaguanine, 5-propyny!cytosine, 5-propynyiuricil, 7- deazaadenine. 7-deaza-8azaadenine, 7-deaza-6-oxopurine, 6-oxopurine, 3-deazaadenosine, 2-oxo-5- methylpyrhnidine, 2-oxo-4-methylthio-5methylp ⁇ yrimidine, 2-thiocarbonyl-4-oxo-5methylpyrimidine,
  • Modified nucieobases can also include abasic moieties.
  • modified nucleotides include biotinylated nucleotides, amine -modifi d nucleotides, alkylated nucleotides, fluorophore-labeled nucleotides, radiolabeled nucleotides, phosphorothioates, phosphoramidites, phosphites, ring atommodified derivatives and the like.
  • a modified nucleotide can be a G-clamp nucleotide.
  • a G-clamp nucleotide refers to a modified cytosine analog wherein the modifications confer the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine nucleotide within a duplex (see, e.g., Lin et al., J Am. Chem. Soc, 120:8531-8532 ( 998); which is incorporated herein by reference in its entirety, in some embodiments, a modified nucleotide can comprise multiple modifications, ⁇ ⁇ some embodiments, a cell can be contacted with any combination of modified nucleotides.
  • the technology described herein relates to a library of siRNA species, the library comprising a plurality of clonal bacterial cell populations; wherein each clonal population comprises bacterial cells as described herein.
  • the bacterial cells can comprise at least a siRNA-binding polypeptide and a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA.
  • a bacterial cell comprises dsRNAs specific for a single target RNA
  • the clonal bacterial population comprising that cell can comprise a population of siRNAs which will specifically bind to the single target RNA and/or which will specifically silence the expression of the target RNA.
  • the technology described herein relates to a library of siRNA species, the library comprising a plurality of populations of siRNAs; wherein each population of siRNAs is obtained according to the methods described herein.
  • a "population of siRNAs" refers to two or more siRNAs, wherein at least two of the siRNAs comprise non-identical sequences, but wherein the two or more siRNAs each comprise a nucleic acid sequence substantially complementary to the same target RNA.
  • a population of siRNAs can comprise two more siRNA species.
  • a population of siRNAs can be generated from a single dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • a population of siRNAs can bind to a single target RNA and/or can specifically silence the expression of the target RNA.
  • a population of siRNAs can be present within a bacterial cell or isolated from a bacterial cell.
  • a bacterial cell library can be in the form of a plurality of multi-well plates, with each well of a plate comprising a clonal bacterial population.
  • the clonal bacterial populations can be provided in media or in glycerol stocks.
  • a library can comprise multiple wells which comprise identical clonal populations, i.e. a clonal population can appear multiple times in a library.
  • a library can comprise a plurality of multi-well plates, with each well of a plate comprising one or more siRNA species (e.g.
  • libraries can be created using automated and/or high-throughput methods, e.g. robotic colony-picking.
  • a library can comprise pooled samples, e.g. multiple clonal bacterial populations, multiple isolated siRNAs, or multiple isolated populations of siRNA species can be pooled so that a smaller number of samples must be initially screened. The individual components of a "positive" pooled can be subsequently screened separately.
  • a library can comprise 10 or more pools of, populations of, and/or individual siRNA species (e.g. isolated or present within bacterial cells), e.g. 10 or more, 100 or more, 1,000 or more, 10,000 or more, or 100,000 or more pools of, populations of, and/or individual siRNA species.
  • siRNA species e.g. isolated or present within bacterial cells
  • a library can comprise a plurality of populations of siRNAs, wherein each population of siRNAs can silence at least one target RNA of a target set.
  • a target set of RNAs can comprise, e.g. the transcriptome of a cell, the transcriptome of an organism, the transcriptome of a cell and/or organism in a specific state (e.g. a diseased organism or an organism at a specific stage of development) or a subtractive transcriptome (e.g. all the transcripts present in a cell under one condition but which are not present in the cell in a second condition).
  • the technology described herein relates to vectors which enable the use of the methods and compositions described herein.
  • the vector can be an expression vector.
  • the vector can be a plasmid.
  • a vector for use in the methods and compositions described herein can comprise (a) a nucleic acid encoding a siRNA-binding polypeptide and (b) a dsRNA cloning site.
  • a dsRNA cloning site further can further comprise a nucleic acid encoding a dsRNA, wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA.
  • a "dsRNA cloning site” refers to a multiple cloning site comprising at least one restriction enzyme site and which can accept the insertion of nucleic acid sequence(s) comprising the sequence of both a sense and anti-sense strand of nucleic acid; wherein one strand is substantially
  • a dsRNA will be encoded and can be expressed, e.g. a sequence inserted at the dsRNA cloning site will be operably linked to a promoter as described herein.
  • a single nucleic acid molecule can comprise the sequence of both the sense and anti-sense strand prior to insertion at the dsRNA cloning site.
  • a dsRNA cloning site can comprise a nucleic acid sequence which comprises sequences which can be cleaved by at least two different restriction enzymes.
  • a dsRNA cloning site can comprise a nucleic acid sequence which comprises sequences which can be cleaved by at least four different restriction enzymes.
  • a dsRNA cloning site can comprise two multiple cloning sites separated by a nucleic acid sequence encoding a hairpin sequence; wherein each multiple cloning site comprises a nucleic acid sequence which comprises sequences which can be cleaved by at least two different restriction enzymes.
  • a vector for use in the methods and/or compositions described herein can comprise at least one constitutive promoter operably linked to at least one of the siRNA- binding polypeptide or the dsRNA multiple cloning site. In some embodiments, a vector for use in the methods and/or compositions described herein can comprise at least one inducible promoter operably linked to at least one of the siRNA-binding polypeptide or the dsRNA multiple cloning site.
  • kits comprising the compositions described herein and kits for practicing the methods described herein.
  • the technology described herein relates to a kit comprising a bacterial cell as described herein, e.g. a bacterial cell comprising at least a siRNA-binding polypeptide and a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA as described herein.
  • a kit for the production of one or more species of siRNA can comprise a vector comprising (a) a nucleic acid encoding a siRNA-binding polypeptide and (b) a dsRNA cloning site.
  • a kit for the production of one or more species of siRNA can comprise two vectors; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and wherein the second vector comprises a dsRNA cloning site.
  • the dsRNA cloning site can further comprise a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially
  • the kit can further comprise a bacterial cell.
  • a kit for the production of one or more species of siRNA can comprise a bacterial cell comprising a siRNA-binding polypeptide and a vector comprising a dsRNA cloning site.
  • the bacterial cell can comprise a nucleic acid encoding a siRNA- binding polypeptide.
  • the nucleic acid encoding a siRNA-binding polypeptide can be a part of an expression vector, a plasmid, a naked nucleic acid, and/or the bacterial genome.
  • the siRNA-binding polypeptide can comprise a purification tag.
  • the siRNA-binding polypeptide can be encoded by a nucleic acid.
  • the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA can be a portion of a vector.
  • at least one of the siRNA-binding polypeptide or the dsRNA can be constitutively expressed.
  • at least one of the siRNA-binding polypeptide or the dsRNA can be inducibly expressed.
  • the bacterial cell can express a siRNA- generating polypeptide.
  • the cell can be an Escherichia coli cell.
  • the technology described herein relates to a kit comprising a library of siRNA species as described herein.
  • compositions and methods described herein can be used to test the efficacy of one or more siRNA species, and/or for the screening of a siRNA library.
  • the efficacy of one or more siRNA species can be assessed in cultured mammalian cells. Methods of targeting mammalian cells with inhibitory RNAs via bacterial invasion are known in the art, see, e.g. Zhao et al. Nature Methods 2005 2:967-973; which is incorporated by reference herein in its entirety.
  • a bacterial cell for use in such an assay can comprise a polypeptide or nucleic acid encoding a polypeptide which can bind to a mammalian cell surface receptor, (e.g. the invasin (inv) gene of Yersinia psuedotuberculosis which binds the integrin receptor of mammalian cells.
  • a bacterial cell for use in such an assay or screen can comprise a mutation reducing the ability of the cell to synthesize or maintain the cell wall, (e.g. deletion of the asd gene of E. coli, thereby rendering the cell a diaminopimelic acid (DAP) auxotroph).
  • DAP diaminopimelic acid
  • Reducing the ability of the cell to synthesize or maintain the cell wall can make the cell susceptible to lysis or degradation after it enters a mammalian cell, thereby releasing inhibitory RNAs (e.g. in the methods described herein, siRNAs) into the mammalian cell.
  • inhibitory RNAs e.g. in the methods described herein, siRNAs
  • the efficacy of one or more siRNA species can be assessed in vivo in C. elegans.
  • dsRNAs readily cross cell membranes in C. elegans, and a number of protocols are known for conducting RNAi in C. elegans, including bacterial feeding assays (see, e.g. Timmons, L., and A. Fire. Nature 1998 395:854 and Lehner et al. Protocol Exchange 2006 159; which are incorporated by reference herein in their entireties.
  • the efficacy of one or more siRNA species can be assessed by first isolating the one or more species of siRNA from the bacterial cells and then contacting a cell and/or organism with the one or more species of siRNA.
  • Methods of introducing ribonucleic acids, and in particular, ribonucleic acids which cause RNAi into various cells and organisms are well known in the art (see, e.g. Sioud, M. "siRNA and miRNA Gene Silencing” Humana Press: 2011 ; “Gene Silencing by RNA Interference” Sohail, M. ed. CRC Press: 2004: each of which are incorporated by reference herein in their entireties).
  • Examples of cells and/or organisms suitable for use in such methods include cultured cells (e.g. mammalian cells or human cells), primary cells, diseased cells (e.g.
  • cancerous cells C. elegans, and Danio rerio.
  • the efficacy of one or more siRNA species can be assessed by screens, selections, and/or by assays.
  • High throughput methods of screening siRNA libraries are known in the art, e.g.
  • siRNA species can be dependent upon the nature of the target RNA, e.g. siRNA species specific for target RNAs which control reproduction in C. elegans can be screened by examining the rate and success of reproduction of worms in the presence of the siRNAs.
  • libraries of siRNA species as described herein, comprising siRNA species targeting a number of different target RNAs can be used in phenotypic screens to identify target RNAs associated with a particular phenotype (e.g. siRNAs which perturb a particular developmental process or which slow the progression of a disease).
  • Phenotypic screens can comprise the assays described above for determining efficacy, e.g. mammalian cell invasion assays.
  • phenotypic screens can involve high-throughput assays.
  • a therapeutic agent comprising a siRNA species or population of siRNA species isolated from a bacterial cell as described herein and/or produced according to the methods described herein.
  • a dsRNA comprising a nucleic acid substantially complementary to a target RNA can be provided to a bacterial cell herein, and a siRNA species and/or population of siRNA species which can be used to reduce the expression of the corresponding target RNA can be produced.
  • target RNAs can be disease-associated RNAs, i.e. RNAs whose overexpression is associated with the cause, progression, or maintenance of a disease state, e.g. oncogenes.
  • target RNAs can be RNAs originating from a pathogenic organism, e.g. the target RNAs can comprise sequences of viral, bacterial, fungal, and/or parasitic origin.
  • target RNAs can be viral RNAs and/or RNAs produced from viral genomic material.
  • a siRNA species and/or population of siRNA species which can be used to reduce the expression of the target RNA can be produced according to the methods described herein and administered to a subject in need of a reduction of the level of expression of the target RNA.
  • a single siRNA species can be administered.
  • a population of siRNA species can be administered.
  • a population of siRNA species can have increased efficacy and a lower likelihood of off-target effects as compared to a single siRNA species.
  • multiple populations of siRNA species can be administered, i.e. multiple target RNAs can be silenced.
  • the technology described herein relates to a pharmaceutical composition comprising a bacterial cell, siRNA species, and/or population of siRNA species according to the methods and compositions described herein.
  • the technology described herein relates to the use of a bacterial cell, siRNA species, or population of siRNA species according to the methods and compositions described herein in the manufacture of a medicament. Methods of preparing medicaments comprising RNA molecules, e.g. siRNAs, are known in the art, (see e.g. Oh and Park. Advanced Drug Delivery Reviews. 2009 61 :850-62; which is incorporated by reference herein in its entirety).
  • the siRNA technology described herein, as well as the methods and compositions relating thereto can be applied to gene silencing applications in any cell and/or organism comprising siRNA machinery.
  • Non-limiting examples include gene silencing applications humans, non-human animals, livestock species, insects (e.g. honeybees), plants, crop plants, etc.
  • the gene silencing can be for therapeutic purposes.
  • the gene silencing can be for agricultural purposes, e.g. to treat agricultural diseases in animals and/or crops or to increase yields in animals and/or crops.
  • a bacterial cell comprising a siRNA-binding polypeptide and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • siRNA-binding polypeptide comprises a
  • siRNA-binding polypeptide selected from the group consisting of:
  • pi 9 polypeptide tombusvirus pi 9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
  • dsRNA comprises nucleic acid sequences substantially complementary to a multiplicity of target RNAs.
  • a method of producing one or more siRNA species which can inhibit the expression of a target RNA comprising:
  • a pharmaceutical composition comprising a siRNA produced according to the method of any of paragraphs 14-17.
  • composition of paragraph 18, further comprising a population of siRNA species comprising a siRNA isolated from a bacterial cell of any of paragraphs 1-13.
  • a vector comprising;
  • nucleic acid encoding a siRNA-binding polypeptide
  • the dsRNA cloning site comprises at least one restriction enzyme site and can accept the insertion of at least one nucleic acid sequence such that a dsRNA is encoded and can be expressed.
  • a vector comprising:
  • nucleic acid encoding a siRNA-binding polypeptide
  • a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • siRNA-binding polypeptide selected from the group consisting of:
  • pi 9 polypeptide tombusvirus pi 9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
  • a library of siRNA species comprising:
  • each clonal population is comprises bacterial cells of any of paragraphs 1-13.
  • a library of siRNA species comprising:
  • each population of siRNAs is obtained according to the methods of any of paragraphs 14-17.
  • each population of siRNAs binds to a single target RNA.
  • a kit comprising a bacterial cell of any of paragraphs 1-13.
  • kits for the production of one or more species of siRNA comprising;
  • a bacterial cell comprising an siRNA-binding polypeptide; and at least one vector comprising a dsRNA cloning site.
  • kits for the production of one or more species of siRNA comprising:
  • a bacterial cell comprising an siRNA-binding polypeptide
  • At least one vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • a kit comprising the vector of any of paragraphs 24-29.
  • kits for the production of one or more species of siRNA comprising two vectors; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide;
  • the second vector comprises a dsRNA cloning site.
  • kits for the production of one or more species of siRNA comprising two plasmids; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide;
  • the second vector comprises a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • the plasmid further comprises a bacterial origin of replication.
  • kits for the production of one or more species of siRNA comprising;
  • a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA cloning site.
  • kits for the production of one or more species of siRNA comprising;
  • a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
  • kits of any of paragraphs 33-47 wherein at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to a constitutive promoter.
  • a kit comprising the library of any of paragraphs 30-32.
  • RNA interference (RNAi) by double-stranded (ds) small interfering RNAs (siRNA) suppresses gene expression by inducing the degradation of mRNAs bearing complementary sequences 1 ' 2 .
  • Endogenous siRNAs imperfectly paired dsRNAs -21-25 nt in length
  • Transfection of synthetic siRNAs into eukaryotic cells 4 ' 5 to silence genes has become an indispensable tool to investigate gene function, and siRNA-based therapy is being developed to knockdown genes implicated in disease 6 .
  • bacteria expressing sense and antisense sequences can be fed to worms to knock down individual genes 7 , no one has used living organisms to produce highly active, purified siRNAs. Described herein is a method to produce highly potent siRNAs from E. coli ectopically expressing pi 9, a siRNA binding protein, which stabilizes siRNA-like species generated by bacterial RNase III.
  • siRNAs are predicted using computer algorithms. siRNAs can also be made from transcribed longer dsRNAs by in vitro biochemical processing by RNase III family enzymes 8 ' 9 . In the latter case, the resulting siRNAs contain many sequences against one target, which sometimes can be more effective than any one sequence 10 , and pools of siRNAs often have fewer off-target effects on genes bearing partially complementary sequences 11 . While gene knockdown by transfection of siRNAs is usually transient, short hairpin RNA construct, delivered by plasmid or lentivirus, is commonly used to achieve stable gene silencing.
  • pl9 an RNAi suppressor protein encoded by the plant RNA virus tombusvirus 12 , selectively binds to -21 nt siRNAs, including those targeting the virus 13 .
  • the pi 9 dimer binds to the -19 nt duplex region of an siRNA in a sequence-independent manner 14 ' 15 . It was originally planned to enrich for endogenous siRNAs in mammalian cells using pi 9 coupled to magnetic beads 15 .
  • pl9 beads were incubated with total RNA isolated from E. coli, an organism that supposedly lacks the RNAi machinery, that was transformed or not with a pcDNA3.1+ plasmid in which pl9 was cloned after the CMV immediate-early promoter.
  • pi 9 protein expression may have stabilized a cryptic siRNA-like RNA species in E. coli.
  • expression of pi 9 also allowed the detection of -21 nt small RNAs ( Figures 5A-5B).
  • siRNA- binding to pi 9 promotes the accumulation of siRNA-like RNAs in E. coli.
  • the nuclease responsible for making small RNAs was sought. The most likely candidate was RNase III, an ancestor of eukaryotic Dicer, responsible for the final step of siRNA biogenesis 17 .
  • coli RNase III is known to generate siRNA-sized dsRNAs from longer dsRNAs in vitro 9 .
  • pl9-expressing plasmids were used to transform two RNase III mutant strains, rncl4 li and rnc38 19 ( Figure ID).
  • pi 9 beads failed to pull down any visible small RNAs.
  • a rncl4 strain also restored the pl9-dependent small RNAs ( Figure IE), providing support for the hypothesis that RNase III is responsible for generating these small RNAs in E. coli.
  • accumulation of these bacterial small RNAs depends on ectopic pi 9 and bacterial RNase III.
  • the GST-pl9-His protein was captured by Nickel (Ni) affinity chromatography and 0.5% SDS was used to selectively elute pl9-bound RNAs that were predominantly -21 nt long ( Figures 2B and 6A-6D). Small RNAs were further purified from other longer RNAs by anion exchange HPLC. To verify that these bacterial small RNAs are double-stranded, they were treated with a variety of nucleases.
  • bacterial small RNAs were sensitive to RNase A, but were insensitive to enzymes that digest ssRNA or DNA (Xrnl, RNase Tl, exonuclease T (Exo T), exonuclease I (Exo I), or DNase Turbo ( Figure 2C).
  • Xrnl, RNase Tl, exonuclease T (Exo T), exonuclease I (Exo I), or DNase Turbo Figure 2C.
  • RNAs that precipitated with anti-Ago were -21 nt long and hybridized to an EGFP probe, but no small RNA precipitated with control mouse IgG. Thus bacterial small RNAs were similar to synthetic siRNA in chemical composition and were incorporated into the RISC. These small RNAs were named 'pro-siRNAs' for prokaryotic siRNAs. [00133] Since pro-siRNAs had properties of siRNAs, whether pl9-captured EGFP pro-siRNAs induce gene knockdown was tested.
  • qRT-PCR and flow cytometry were used to compare mRNA and protein knockdown, respectively, of dlEGFP in HeLa-dlEGFP cells transfected with a synthetic EGFP siRNA or pro-siRNAs purified from E. coli expressing pi 9 and hairpins of either full length EGFP (EGFPFL) or a 100 nt fragment that overlapped with the EGFP siRNA sequence (EGFP 100).
  • EGFPFL and EGFP 100 pro-siRNAs knocked down EGFP expression more effectively than equimolar concentrations of siRNA ( Figures 2E and 7A).
  • pro-siRNAs made from the plasmid without or with only half of the EGFP hairpin could not silence EGFP effectively ( Figure 7B).
  • LMNA which encodes two splice variant products, lamin A and lamin C
  • PLKl encodes two splice variant products, lamin A and lamin C
  • HIV vif viral infectivity factor
  • gag gag antigen
  • the HPLC-purified pro-siRNAs for each gene contained a few different sized species that migrated close to the 21 nt marker on both native and denaturing polyacrylamide gels (Figure 2F).
  • LMNA and PLKl pro-siRNAs a minor RNA band migrated at -25 nt.
  • LMNA and TP53 siRNAs were from a single sequence; PLKl siRNAs were a pool of 4 siRNAs and were chemically modified by proprietary methods for enhanced stability and reduced off-target effects 21 ) in HeLa-dlEGFP and HCT116 cells were compared.
  • pro-siRNAs dose response experiments comparing transfection of pro- siRNAs (0.2, 2, 20 nM) targeting LMNA, TP53 and PLKl with five commercial siRNAs for each gene (four siRNAs from Dharmacon, of which the PLKl siRNAs were chemically modified for enhanced RISC uptake or stability by proprietary methods, and one siRNA sequence chosen based on published effectiveness) were performed (Figure 9).
  • the potency of the commercial siRNAs varied, as best evaluated at the lowest concentration.
  • the pro-siRNAs whose sequences were not optimized, achieved similar gene knockdown as the commercially optimized siRNAs. At a concentration of 2 nM, each pro-siRNA achieved knockdown of -90%.
  • siRNAs and pro-siRNAs targeting vif ' did not alter the percentage of initially infected HeLa-CD4 cells (data not shown), but did suppress vif gene expression and inhibit subsequent rounds of infection, assessed in the TZM-bl luciferase reporter cell line ( Figure 3D).
  • Transfection of vif pro-siRNAs resulted in much lower levels of vz/mRNA in HeLa-CD4 cells and HIV tat-driven luciferase activity, compared to transfection with either or both vz/siRNAs.
  • pro-siRNAs were superior to previously used siRNAs in inhibiting HIV spread in vitro.
  • RNAi RNAi to suppress HIV or other viruses
  • sequence diversity Because pro-siRNAs target many sequences within a gene, pro-siRNAs directed against a viral gene can have broader activity against diverse viral strains than siRNAs and can also be less likely to generate siRNA-resistant mutants.
  • gag pro-siRNAs were engineered using hairpins with 200 and 500 nt long stems from the gag coding region of clade B HIV-III B virus.
  • the gagB200 and gagB500 pro-siRNAs more potently suppressed HIV-III B , than the previous gag siRNA ( Figure 2D).
  • both gag pro-siRNAs knocked down gag mRNA and inhibited viral spread in vitro for UG29 (clade A) and ⁇ 22 (clade C) viruses, although they worked slightly less effectively than against III B virus.
  • pro-siRNAs are contaminated with endotoxin. Although SDS-eluted pro-siRNAs contained significant amounts of endotoxin, assayed by Limulus amoebocyte lysate (LAL) assay, HPLC purified pro-siRNAs, even at concentrations as high as 320 nM, were below the limit of detection (0.25 EU/ml) (Table 1).
  • LAL Limulus amoebocyte lysate
  • Endotoxin contamination was tested for by assaying for induction of mRNA expression of the proinflammatory cytokines TNFA, IL6, IL8 and IL12, measured 4 hr later by qRT-PCR in highly endotoxin-sensitive monocyte-derived human macrophages (Figure 10A).
  • Incubation with HPLC- purified vz/pro-siRNAs (320 nM) did not trigger cytokine gene expression.
  • pro-siRNAs did not contain significant amounts of immunostimulatory endotoxin.
  • Next MDMs were transfected with a few siRNAs and pro-siRNAs (at 20 nM) to test immune response mediated by endogenous immune sensors (Figure 10B).
  • siRNA and pro-siRNA against LMNA efficiently down regulated LMNA rriRNA, indicating these siRNAs were successfully transfected into MDMs. Comparing to siRNAs, pro-siRNAs did not trigger excessive activation of immune genes.
  • pro-siRNAs were cloned and deep sequenced using a cloning method established for eukaryotic siRNAs (sequencing reads and alignment summary in Table 2). Most reads were concentrated between 20 and 22 nt ( Figures 4A and 11). The majority of reads (on average -75%) aligned to the target sequence, plasmid backbone or the E. coli genome. The vast majority of aligned sequences (82-99%) originated from the target sequence ( Figure 4B); consistent with the efficient gene knockdown they induced. Reads were generated from the entire target sequence, but were also concentrated at specific sites ('hot spots') ( Figures 4C, 11, and 12A-E).
  • pro-siRNAs are mostly dsRNAs and the strand bias in the deep sequencing data likely reflects ligation bias during cloning.
  • coli RNase III might process dsRNA into siRNA-sized small RNAs in vivo through a mechanism that differs from Dicer 27 , whose cleavage of a long dsRNA results in phased and evenly distributed sequences along a target gene.
  • pro-siRNAs contained non-targeting sequences derived from the plasmid or E. coli genome, possible off-targeting effects 26 were investigated.
  • RNA expression profiles were compared by RNA deep sequencing of HeLa-dlEGFP cells transfected with 4 nM of negative control or EGFP siRNA or EGFPFL or EGFPIOO pro-siRNAs (sequencing reads and alignment summary in Table 2). Tophat and Cufflinks were used to analyze the data and plotted volcano plots of all annotated transcripts (fold change versus p value, Figure 4D).
  • EGFPIOO pro-siRNA had higher number of significantly changed genes while EGFPFL pro-siRNA had less ( Figures 4F and 14A).
  • EGFPFL pro-siRNA also produced the least changes in long non-coding RNAs, a group of newly discovered gene regulators ( Figures 14B-14C).
  • EGFPIOO pro-siRNAs made from a shorter hairpin (100 bp), contained higher proportion of plasmid and genomic sequences compared to other pro-siRNAs made from longer hairpins (200 to 720 bp, Figure 4B), which is likely the cause of higher off-target effect. These data indicate a plasmid containing longer sequences of the target gene could have fewer off-target effects.
  • LMNA pro-siRNAs were also compared by microarray. Consistent with the EGFP data, LMNA pro-siRNAs, made from a longer hairpin (523 bp), produced fewer number of significantly changed genes comparing to LMNA siRNA ( Figures 4E, 4F, and 14D). The RNA profiling data also showed the target gene was always the most down regulated gene and pro-siRNAs consistently produced better knockdown than siRNA. Thus pro-siRNAs could be engineered to offer better knockdown and lower off-target effects compared to synthetic siRNAs.
  • pro- siRNAs offer highly specific knockdown that is at least as good as synthetic siRNAs without the need to test multiple sequences.
  • bacteria can be genetically engineered to produce siRNAs that are highly effective and not toxic to mammalian cells. Specifically, it demonstrated herein is efficient knockdown of one exogenous gene ⁇ EGFP), two viral genes (vif and gag) and 3 host genes (PLKl, TP53, LMNA).
  • ⁇ EGFP exogenous gene
  • vif and gag viral genes
  • PLKl TP53
  • LMNA 3 host genes
  • pro-siRNAs are natural products of RNase III, they likely have favorable ends (e. g., 5'-phosphate, 3'-hydroxyl and 3' overhangs) for efficient loading by Ago into the RISC and do not activate cytosolic innate immune RNA sensors.
  • pro-siRNAs for research purposes might be more cost effective than purchasing and testing multiple individual chemically synthesized siRNAs.
  • pro-siRNAs containing multiple sequences, might offer fewer off-target effects than individual siRNAs and could be harder for the target gene to escape silencing by mutation.
  • chemical synthesis provides the opportunity for chemical modifications to increase potency, enhance stability and reduce off-target effects or couple fluorophores or targeting moieties. Such modifications might also be possible for pro-siRNAs, either by adding modified ribonucleotides to bacterial cultures during IPTG induction or by performing the same coupling reactions with purified pro-siRNAs as are used to modify siRNAs, respectively.
  • RNase Ill-deficient E. coli expressing dsRNAs can be fed to C. elegans li and bacteria- derived dsRNAs can be applied to plants to induce specific gene knockdown 28 .
  • gene silencing requires host Dicer and, unlike for mammalian cells, is enhanced in these organisms by RNA-dependent RNA polymerases that can amplify small amounts of RNA.
  • genetically engineered E. coli designed to express an invasin to induce bacterial uptake and listeriolysin, to allow bacterial RNAs to escape from phagolysosomes, delivered dsRNAs into the cytoplasm of human cells through "trans-kingdom RNAi" technology 29 .
  • pro-siRNAs described here, could become a valuable cost effective addition to existing RNAi techniques for both research and therapeutics.
  • the method described herein for producing pro- siRNAs can easily be adopted and scaled-up in an industrial setting. It is contemplated that mammalian cDNA libraries could be used to generate pro-siRNA libraries, e.g. for siRNA screening pro-siRNAs, generated from longer hairpins containing multiple sequences, might offer fewer off- target effects than individual siRNAs and in the cases of virus infection or cancer might be harder for the target gene to escape from by mutation.
  • chemical synthesis provides the opportunity for chemical modifications to increase potency, enhance stability and reduce off-target effects or to couple fluorophores or targeting moieties.
  • pro- siRNAs e.g. either by adding modified ribonucleotides to bacterial cultures during IPTG induction or by performing the same coupling reactions with purified pro-siRNAs as are used to modify siRNAs, respectively.
  • E. coli strain DH5a was used for cloning and for initial characterization of the siRNA-like RNA species.
  • T7 Express Iq NEB
  • rnc- 74::DTnlO Tet R
  • Ornc-38 Kan R
  • coli strain MG1655 AlacZYA also referred as MG1655 Mac. All E. coli strains were cultured in LB broth, Lennox (BD) at 37°C with shaking at 250 rpm and antibiotics when required were used at the following concentrations; carbenicillin (100 ⁇ g/ml), kanamycin (50 ⁇ g/ml), spectinomycin (50 ⁇ g/ml), tetracycline (12.5 ⁇ g/ml).
  • Listeria monocytogenes strain 10403S was cultured in brain-heart infusion medium (BD Biosciences) at 30°C. Transformation of bacterial cells was performed as previously described 32 .
  • pLIV-l-His-pl9 plasmid was used, which encodes pi 9 with an N-terminal His tag cloned in pLIV-1 plasmid (gift of Spotify Higgins, Harvard Medical School).
  • E. coli RNase III with an N-terminal FLAG was cloned in pcDNA3.1+ and pCDF- lb (Novagen) plasmids.
  • the hairpin RNA sequences were: EGFPFL, the entire 720-bp EGFP coding sequence (from pEGFP-Nl, Clontech); EGFP100, a 100 bp from nt 219 to 318; EGFP Hotspot-1 360 bp from nt 1 to 360; EGFP Hotspot-2 360 bp from nt 361 to 720; LMNA (NM 005572.3), 523 bp from nt 267 to 789; TP53 (NM_000546.5), 301 bp from nt 376 to 676; PLK1 (NM_005030.3), 299 bp from nt 92 to 390; vz/(K03455), the entire 579-bp; gag (K03455), gagB200: 200 bp from nt 1183 to 1382, gagB500 500 bp from nt 1004 to 1503. (Genbank entries listed; numbers refer to position with respect to the
  • the GST-pl9-His protein was cloned under the control of the T7 promoter in pRSF-lb (Novagen) or pCDF-lb to generate pRSF-GST-pl9-His and pCDF-GST-pl9-His.
  • the second plasmid is a L4440 plasmid encoding the entire EGFP coding sequence (L4440-EGFP).
  • U87.CD4.CXCR4 and U87.CD4.CCR5 cells were cultured in DMEM medium (Invitrogen) supplemented with 10% heat-inactivated fetal bovine serum (FBS).
  • ACH2 cells human leukemia T cell line CEM latently infected with HIV-1 were cultured in RPMI medium (Invitrogen)
  • monocytes were isolated from blood of a healthy donor by Ficoll-Paque Plus (GE Healthcare) density separation. Monocytes were plated on
  • PRIMARIA plates FALCON
  • RPMI medium Invitrogen
  • 10% heat- inactivated human serum and adherent cells were cultured for 5 d to allow differentiation into macrophages.
  • RNA isolation and qRT-PCR Total RNA was isolated from 3 ml of E. coli stationary phase culture with 1 ml Trizol reagent (Invitrogen) following the manufacturer's protocol. RNA from human cells was collected in Trizol and extracted according to the manufacturer's protocol. Total RNA (1 ⁇ g) was converted to cDNA using Superscript III Reverse Transcriptase (Invitrogen). For qRT-PCR, 10 ⁇ reaction, containing SsoFast EvaGreen mastermix (Bio-Rad), appropriate primers (Table 4), and template cDNAs made from 10 ng RNA, was amplified on a Bio-Rad CFX 96 Thermal Cycler. All qRT-PCR data were normalized to the human GAPDH gene. qRT-PCR primers for human genes (Table 6) were selected from PrimerBank (available on the world wide web at
  • siRNA isolation from total RNA using pl9 magnetic beads were prepared at NEB as previously described 15 .
  • To pull down siRNAs 50 ⁇ g of total RNA (isolated from human or E. coli cells) was used following the manufacturer's protocol 15 .
  • GST-pl9-His was purified as follows. A fresh single transformant of T7 Express Iq containing pGEX-4T-l -pl9-T7 was used to inoculate 300 ml LB medium in a 1.5 L flask. When the OD 60 o reached 0.3-0.6, protein and pro- siRNA expression were induced by adding 0.5 mM IPTG for 1 hr.
  • HPLC buffers were: Buffer A, 25 mM Tris-HCl, 2 mM EDTA; Buffer B, 25mM Tris-HCl, 2 mM EDTA, 5 M NaCl.
  • HPLC was initiated with a flow rate of 1 ml/min at 25°C. Elution was performed using a linear gradient of 0-10% Buffer B over 4 min, followed by 10% Buffer B for 6 min, and a second linear gradient of 10-25%) Buffer B over 15 min at a reduced flow rate of 0.5 ml/min. pro-siRNA eluted in the second gradient was collected by isopropanol precipitation.
  • RNA size standards (miRNA marker, siRNA marker and Low Range ssRNA Ladder) were from NEB.
  • RNA samples were prepared in Gel Loading Buffer II (Ambion) without heat denaturation and electrophoresis was performed at room temperature.
  • Nuclease sensitivity assay The nucleases tested were: RNase A, RNase Tl, and Turbo DNase (all from Ambion), Xrnl, exonuclease T, and exonuclease I (all from NEB).
  • RNase A RNase A
  • RNase Tl RNase Tl
  • Turbo DNase All from Ambion
  • Xrnl exonuclease T
  • exonuclease I all from NEB.
  • 200 ng of an unmodified synthetic negative control siRNA (GenePharma) and vz/pro-siRNA were used and assays were incubated in a 20 ⁇ reaction volume using standard amounts of enzymes at 37°C for 1 hr.
  • Treated RNAs were purified by phenol/chloroform extraction followed by isopropanol precipitation.
  • LPS Lipopolysaccharide
  • MDM plated in 24 well plates were incubated with medium containing RNA or LPS at the indicated concentration for 4 hr before harvesting RNA.
  • siRNAs and pro-siRNAs were also transfected to MDMs at 20 nM using
  • Lipofectamine 2000 (Invitrogen) and total RNA were harvest at 24hrs after transfection.
  • RNA samples were dephosphorylated by Antarctic Phosphatase (NEB) for 30 min at 37°C in the presence of Murine RNase Inhibitor (NEB).
  • NEB Antarctic Phosphatase
  • RNA northern blot Small RNA northern blot.
  • Northern blot for small RNAs was performed as previously described 33 .
  • the EGFP specific sense probe was a 32 P-UTP -internally labeled RNA prepared by in vitro transcription using T7 RNA polymerase (NEB) and a PCR-generated DNA template of the full- length EGFP gene that incorporated a T7 promoter.
  • NEB T7 RNA polymerase
  • siRNA transfection for testing RNA silencing efficiency. All siRNA transfections were performed using Lipofectamine 2000 following the manufacturer's protocol. Briefly, cells were plated in 24 well plates ( 1 x 10 5 per well) and the transfection complex (containing 1.0 ml Lipofectamine 2000 and siRNAs) was added directly to the medium. RNA and protein samples were isolated from cells 24 hr post-transfection. For the PLK1 cell killing experiment, cells were counted using a TC-10 automatic cell counter (Bio-Rad).
  • siRNAs ON-TARGETplus Non-targeting siRNA #4 (D-001810-04-05, Dharmacon), siGENOME Lamin A/C Control siRNA (D-001050-01-20, Dharmacon), Set of 4: siGENOME LMNA siRNA (MQ-004978-01 -0002, Dharmacon), ON- TARGETplus SMARTpool - Human PLK1 (L-003290-00-0005, Dharmacon), Set of 4 Upgrade: ON- TARGETplus PLK1 siRNA(LU-003290-00-0002, Dharmacon), Set of 4: siGENOME TP53 siRNA (MQ-003329-03-0002, Dharmacon), Negative control siRNA (NC siRNA, B01001, GenePharma), Positive control siRNA TP53 (B03001, GenePharma), custom EGFP siRNA (sense,
  • AAUCAGCACCUGCCAUCUGTT SEQ ID NO: 119
  • custom gag siRNA (sense
  • RISC Immunoprecipitation Cells (3x10 6 ) were transfected with 4 nM NC siRNA or EGFPFL pro-siRNAs. After 24 hours cells were scraped from the plate in 2 ml lysis buffer (150 mM KC1, 25 mM Tris-HCl pH 7.5, 2 mM EDTA, 0.5 mM DTT, 1% NP-40 and Roche Complete Protease Inhibitor Cocktail). Cells were then mechanically disrupted for 1 min using a micro-MiniBeadbeater (BioSpec). The cell lysate was incubated at 4°C with rotation for 1 hr to ensure complete lysis.
  • 2 ml lysis buffer 150 mM KC1, 25 mM Tris-HCl pH 7.5, 2 mM EDTA, 0.5 mM DTT, 1% NP-40 and Roche Complete Protease Inhibitor Cocktail. Cells were then mechanically disrupted for 1 min using a micro-MiniBeadbeat
  • IP was performed by adding anti-Ago (2A8) antibody (Millipore, MABE56) or mouse total IgG (Jackson Labs) at 1 : 100 dilution together with 30 ⁇ protein G Dynabeads (Invitrogen) and samples were rotated at 4°C overnight. After washing 4 times in lysis buffer, precipitated RNAs were isolated using Trizol reagent from 90% of the reaction mix, while 10% was saved for immunoblot input.
  • Antibodies and their dilutions were: anti-FLAG (M2) 1 : 1,000 (Sigma- Aldrich, F1804), anti-His tag 1 :500 (Covance, MMS-156P), anti-PLKl 1 : 100 (Santa Cruz, sc-17783), anti-LaminA/C 1 : 1,000 (Santa Cruz, sc-7292), anti-p53 (DO-1) 1 :500, (Santa Cruz, sc-126), anti-beta-Tubulin 1 : 10,000 (Sigma- Aldrich, T5168), anti-Ago (2A8) 1 : 1,000 (Millipore, MABE56).
  • Horseradish peroxidase conjugated anti-mouse or anti-rabbit IgG secondary antibodies were used at 1 :5,000 dilution followed by incubating the membranes in SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific).
  • DNA oligonucleotides purchased from IDT were PAGE purified. Purified DNA oligonucleotides (10 pmol) were end-labeled with ⁇ - 32 ⁇ ATP by T4 Polynucleotide Kinase (NEB) and 2 pmol was then mixed with 5 ng of pro- siRNAs in buffer containing 20 mM Tris-HCl pH 7.9, 100 mM NaCl and 2 mM EDTA. Samples were heated to 80°C for 10 min and allowed to cool to room temperature. A fraction of the sample was separated on a native 15% polyacrylamide gel. The gel was directly exposed to a phosphorimager screen. Multi-gauge software (Fujifilm) was used for image quantification.
  • NEB Polynucleotide Kinase
  • siRNA library preparation deep sequencing, and data analysis.
  • siRNAs were cloned according to the Illumina small RNA sample preparation guide vl .5 with the following exceptions.
  • Custom 5' RNA ligation adapters were synthesized with a 4 nt nucleotide barcode sequence (Table 7).
  • Small RNA libraries were pooled and sequenced on one sequencing lane of an Illumina GAII sequencer (Genome Technology Core, Whitehead Institute or NEB). Novocraft software
  • rriRNA profiling by microarray and deep sequencing siRNAs and pro-siRNAs (4 tiM) were transfected into HeLa-dlEGFP cells and RNA was isolated 24 hr post-transfection. Non- targeting siRNA #4 (Dharmacon) was used as negative control siRNA. Data from biological duplicates were analyzed at the Microarray Core, Dana Farber Cancer Institute for microarray analysis using GeneChip 1.0 ST (Affymetrix). Microarray data was analyzed using dChip software and p values of gene expression changes were calculated using paired T-test method 34 . Original data and analysis files are available upon request.
  • RNA deep sequencing Ribo-Zero rRNA Removal Kits (Epicentre) was used to remove large ribosomal RNAs from total RNA following the manufacturer's protocol. rRNA-depleted RNA (from 500 ng total RNA) was used to construct deep sequencing library using NEBNext Ultra RNA Library Prep Kit for Illumina (NEB #E7530) according to the manufacturer's protocol. Illumina GAII was used for sequencing (NEB). Tophat and Cufflinks software suites were used to analyzed the RNA deep sequencing data from biological duplicates. Reference genome was Human genome GRCh37/hgl9 and annotations of lincRNA transcripts were downloaded from UCSD genome browser. Original data and analysis files are available upon request.
  • Flow cytometry For EGFP, cells were removed from plates by trypsin digestion and re- suspended in FACS buffer, DPBS (Invitrogen) containing 2% heat-inactivated FBS. Intracellular staining of p24 antigen was performed using an Intracellular Staining Kit (Invitrogen) according to the manufacturer's protocol and fluorescein-labeled p24 antibody (1 :200, Beckman Coulter, cat#KC57-FITC). Fluorescence was analyzed on a FACSCalibur (BD) using FlowJo software (Tree Star).
  • HeLa-CD4 cells were transfected with 4 nM siRNA and pro-siRNA in 24 well plates ( 1 x 10 5 cells/well). Cells were infected 12 hr post-transfection with HIVIIIB (-400 ng/ml p24) and culture medium was changed 12 hr post-infection.
  • HIV UG29 U87.CD4.CXCR4 cells were used and for HIV IN22 U87.CD4.CCR5 cells were used.
  • Culture medium was collected for TZM-bl assay and RNA was extracted for qRT-PCR 24-36 hr post- infection.
  • TZM-bl cells plated in 24 well plates (l x lO 5 cells/well) 12 hr before, were analyzed 24 later by luciferase assay performed using a Luciferase Assay System kit (Promega) following the manufacturer's protocol.
  • Silhavy D. et al. A viral protein suppresses RNA silencing and binds silencing-generated, 21- to 25-nucleotide double-stranded RNAs. Embo J 21, 3070-3080 (2002). Vargason, J.M., Szittya, G., Burgyan, J. & Hall, T.M. Size selective recognition of siRNA by an RNA silencing suppressor. Cell 115, 799-811 (2003).
  • pGEX-4T-l-P19- For expression of GST-P19-His from Tac promoter, and T7 hairpin RNA from the T7 promoter
  • Gag200-RSalI ATCGTCGACTCTTCTGGTGGGGCTG 65 pGEX-4T-l -P19- TTGGCT T7-GagB200
  • IL12 For CACTCCCAAAACCTGCTGCTGAG 74 qRT-PCR IL12 Rev TCTCTTCAGAAGTGCAAGGGTA 75 qRT-PCR IL6 For GATGAGTACAAAAGTCCTGATCCA 76 qRT-PCR IL6 Rev CTGCAGCCACTGGTTCTGT 77 qRT-PCR IL8 For AGACAGCAGAGCACACAAGC 78 qRT-PCR IL8 Rev ATGGTTCCTTCCGGTGGT 79 qRT-PCR TNFA For CAGCCTCTTCTCCTTCCTGAT 80 qRT-PCR TNFA Rev GCCAGAGGGCTGATTAGAGA 81 qRT-PCR Vif For AGGGAAAGCTAGGGGATGGTTTT 82 qRT-PCR Vif Rev CCCAAATGCCAGTCTCTTTCTCC 83 qRT-PCR TN22-Vif For AAAGAGAGCTAATGGATGGTTTTTT 84 qRT-PCR IN22-Vif Rev CCCAAATGCCAAT

Abstract

The technology described herein relates to siRNAs, e.g., methods and compositions relating to the production of siRNAs in bacterial cells.

Description

METHODS AND COMPOSITIONS FOR THE PRODUCTION OF SIRNAS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 61/751,489 filed January 11, 2013, the contents of which are incorporated herein by reference in their entirety.
GOVERNMENT SUPPORT
[0002] This invention was made with federal funding under Grant Nos. AI087431 awarded by the National Institutes of Health. The U.S. government has certain rights in the invention.
SEQUENCE LISTING
[0003] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on January 9, 2014, is named 701039-075001-PCT_SL.txt and is 33,506 bytes in size.
TECHNICAL FIELD
[0004] The technology described herein relates to methods of producing siRNAs in vivo in bacterial cells.
BACKGROUND
[0005] RNA interference (RNAi) by double-stranded (ds) small interfering RNAs (siRNA) suppresses gene expression by inducing the degradation of mRNAs bearing complementary sequences (Fire, A. et al. Nature 1998 391 :806-811 ; Hamilton, A.J. & Baulcombe, D.C. Science 1999 286:950- 952). Transfection of synthetic siRNAs into eukaryotic cells to silence genes has become an indispensable tool to investigate gene function, and siRNA-based therapy is being developed to knockdown genes implicated in disease (Elbashir, S.M. et al. Nature 2001, 411 :494-8; Caplen, N.J., et al. Proc Natl Acad Sci USA 2001 98:97427; Rettig, G.R. & Behlke, M.A. Mol Ther 2012 20:483- 512). More efficient ways to produce siRNAs are desired.
SUMMARY
[0006] The technology described herein is directed to methods and compositions relating to the production of active siRNAs generated in vivo, e.g. in bacterial cells.
[0007] In one aspect, described herein is a bacterial cell comprising a siRNA-binding polypeptide and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. In some embodiments, the siRNA-binding polypeptide comprises a purification tag. In some embodiments, the siRNA-binding polypeptide is encoded by a nucleic acid. In some embodiments, the siRNA-binding polypeptide is selected from the group consisting of: pi 9 polypeptide; tombusvirus pl9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pi 30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide. In some embodiments, the dsRNA is greater than 21 nucleotides in length. In some embodiments, the dsRNA is a hairpin RNA. In some embodiments, the bacterial cell expresses an RNase III polypeptide. In some embodiments, the bacterial cell expresses an RNase III polypeptide encoded by an exogenous nucleic acid sequence. In some embodiments, the bacterial cell is an Escherichia coli cell. In some embodiments, at least one of the siRNA-binding polypeptide and the dsRNA are constitutively expressed. In some embodiments, at least one of the siRNA-binding polypeptide and the dsRNA are inducibly expressed. In some embodiments, the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA is part of a plasmid.
[0008] In one aspect, described herein is a method of producing one or more siRNA species which can inhibit the expression of a target RNA, the method comprising: culturing a bacterial cell as described herein under conditions suitable for the production of siRNAs. In some embodiments, the method further comprises a second step of isolating the siRNA-binding polypeptide and eluting the siRNAs bound to the siRNA-binding polypeptide. In some embodiments, the method further comprises purifying the siRNAs eluted from the siRNA-binding polypeptide by chromatography e.g. anion exchange HPLC. In some embodiments, the method further comprises contacting the cell with one or more modified nucleotides before or during the culturing step.
[0009] In one aspect, described herein is a pharmaceutical composition comprising a siRNA produced according to the methods described herein. In some embodiments, the composition further comprises a population of siRNA species.
[0010] In one aspect, described herein is a pharmaceutical composition comprising a siRNA isolated from a bacterial cell as described herein. In some embodiments, the composition further comprises a population of siRNA species.
[0011] In one aspect, described herein is the use of a siRNA produced according to the methods described herein in the production of a medicament.
[0012] In one aspect, described herein is the use of a siRNA isolated from a bacterial cell of as described herein in the production of a medicament.
[0013] In one aspect, described herein is a vector comprising; a nucleic acid encoding a siRNA- binding polypeptide; and a dsRNA cloning site. In some embodiments, the dsRNA cloning site comprises at least one restriction enzyme site and can accept the insertion of at least one nucleic acid sequence such that a dsRNA is encoded and can be expressed. In one aspect, described herein is a vector comprising: a nucleic acid encoding a siRNA-binding polypeptide; and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. In some embodiments, the siRNA-binding polypeptide is selected from the group consisting of: pi 9 polypeptide; tombusvirus pl9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide. In some embodiments, the vector is a plasmid. In some embodiments, the plasmid further comprises a bacterial origin of replication.
[0014] In one aspect, described herein is a library of siRNA species, the library comprising: a plurality of clonal bacterial cell populations; wherein each clonal population is comprises bacterial cells as described herein. In one aspect, described herein is a library of siRNA species, the library comprising: a plurality of populations of siRNAs; wherein each population of siRNAs is obtained according to the methods described herein. In some embodiments, each population of siRNAs binds to a single target RNA.
[0015] In one aspect, described herein is a kit comprising a bacterial cell as described herein. In one aspect, described herein is a kit for the production of one or more species of siRNA, the kit comprising: a bacterial cell comprising an siRNA-binding polypeptide; and at least one vector comprising a dsRNA cloning site. In one aspect, described herein is a kit for the production of one or more species of siRNA, the kit comprising: a bacterial cell comprising an siRNA-binding
polypeptide; and at least one vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. In one aspect, described herein is a kit comprising a vector as described herein. In one aspect, described herein is a kit for the production of one or more species of siRNA, the kit comprising two vectors; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and wherein the second vector comprises a dsRNA cloning site. In one aspect, described herein is a kit for the production of one or more species of siRNA, the kit comprising two plasmids; wherein the first vector comprises a nucleic acid encoding a siRNA- binding polypeptide; and wherein the second vector comprises a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. In some embodiments, at least one vector is a plasmid. In some embodiments, the plasmid further comprises a bacterial origin of replication. In some embodiments, the kit further comprises a bacterial cell. In one aspect, described herein is a kit for the production of one or more species of siRNA, the kit comprising; a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA cloning site. In one aspect, described herein is a kit for the production of one or more species of siRNA, the kit comprising; a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. In some embodiments, the siRNA-binding polypeptide comprises a purification tag. In some embodiments, the siRNA-binding polypeptide is encoded by a nucleic acid. In some embodiments, the bacterial cell expresses an RNase III polypeptide. In some embodiments, the cell is an Escherichia coli cell. In some embodiments, at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to a constitutive promoter. In some embodiments, at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to an inducible promoter. In some embodiments, the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA is part of a plasmid. In one aspect, described herein is a kit comprising the library as described herein. BRIEF DESCRIPTION OF THE DRAWINGS
[0016] Figures 1A-1E demonstrate that ectopic pl9 expression captures small RNAs in E. coli. Figure 1A depicts an image of a gel from experiments in which pl9-coupled magnetic beads were used to isolate small dsRNAs from total RNA isolated from mammalian ACH2 cells, or from E. coli cells, or from E. coli cells containing a pcDNA3.1-P19 expression plasmid. Captured RNAs were 5' 32P-labeled, separated on a native polyacrylamide gel and detected by autoradiography. Figure IB depicts images of gels from experiments in which expression of FLAG-tagged pi 9, but not TREX1 or empty plasmid (V, vector), from pcDNA3.1+ in E. coli led to accumulation of -21 nt RNAs. Total RNAs purified from E. coli containing an empty vector, or pcDNA3.1+ expressing FLAG-tagged pi 9 or TREXl were separated on a denaturing polyacrylamide gel and stained with SYBR Gold. FLAG immunoblot verified protein expression. Figure 1C depicts an image of a gel from experiments in which total RNAs purified from E. coli containing an empty vector, or pcDNA3.1+ expressing His- tagged pi 9 or His-tagged pl9 mutant proteins defective in RNA binding (Mutl 14 : W39G, W42G and Mut216: K71A, R72G) were separated on a denaturing polyacrylamide gel and stained with SYBR Gold. His immunoblot verified protein expression. Figure ID depicts images of gels from experiments in which pl9-associated RNAs were isolated with pl9-coupled magnetic beads from total RNA extracted from WT E. coli (DH5a or MG1655 Alac) or RNase III mutant strains (in MG1655 Alac background) expressing the His-tagged pi 9 protein. pl9-captured RNAs were separated on native or denaturing gels and stained with SYBR Gold, pi 9 expression was verified by His immunoblot. The asterisk (*) indicates equal loading of a background band. Figure IE depicts an image of a gel from experiments in which pl9-associated RNAs were isolated with pl9-coupled magnetic beads from total RNA extracted from pl9 expressing E. coli WT BL21(DE3) cells or rncl4 mutant HTl 15(DE3) cells co-transfected with pi 9 and pcDNA3.1 vector that was empty or encoded Flag-tagged E. coli RNase III. pl9-captured RNAs were separated on a native polyacrylamide gel and stained with SYBR Gold, pi 9 and RNase III expression was verified by immunoblots. M, markers. Arrows indicate the -21 nt small RNA band. Data are representative of at least 2 independent experiments.
[0017] Figures 2A-2F demonstrate pro-siRNAs knockdown EGFP expression. Figure 2A depicts a schematic of pGEX-4T-l -pl9-T7 plasmid and the method to produce pro-siRNAs from E. coli. Figure 2B depict an image of a gel from experiments in which anion exchange HPLC fractions of SDS-eluted RNAs (isolated from E. coli transformed to express pro-siRNAs) were separated on a native polyacrylamide gel and stained with SYBR Gold. Figure 2C depicts images of gels from experiments in which nuclease sensitivity assay confirms pro-siRNAs are dsRNAs. Synthetic siRNAs and HPLC purified pro-siRNAs were incubated with nucleases and separated on a native polyacrylamide gel stained with SYBR Gold. Figure 2D depicts images of gels from experiments in which anti-Ago mAb 2A8 or mouse total IgG was used to immunoprecipitate RNAs in negative control (NC) siRNA or EGFPFL pro-siRNA-transfected HeLa-dlEGFP cells. Immunoprecipitated RNAs were analyzed by Northern blot using a probe complementary to the full length EGFP coding sequence (top) or 5' 32P end-labeling (middle). Bottom immunoblot is probed for Ago protein. Figure 2E depicts a graph of the results of qRT-PCR of EGFP expression and EGFP mean fluorescence intensity by flow cytometry in HeLa-dlEGFP cells transfected with either 4 nM of siRNA or pro- siRNAs. Data are normalized to cells treated with negative control (NC) siRNA and are mean±SD of 2 (qRT-PCR) and 3 (EGFP fluorescence) independent experiments. mRNA level is relative to GADPH. Figure 2F depicts images og gels from experiments in which HPLC-purified pro-siRNAs were separated on native or denaturing polyacrylamide gels stained with SYBR Gold.
[0018] Figures 3A-3D demonstrate that pro-siRNA-mediated knockdown of endogenous and viral gene expression in human cells. Figure 3A depicts graphs and images of gels from experiments in which qRT-PCR and immunoblot assays of genes targeted for knockdown with the indicated siRNAs or pro-siRNAs, which were transfected (4 nM) into HeLa-dlEGFP (top) or HCT116 (bottom) cells. Figure 3B depicts graphs of cell counts after transfection with PLK1 siRNA or pro-siRNA (4 nM) or negative control (NC) siRNA or EGFP pro-siRNA as nontargeting controls, respectively. Figure 3C depicts a schematic and graphs of the results of experiments demonstrating inhibition of HIV-1 spreading by transfection of vif siRNAs and pro-siRNAs (4 nM). vif siRNAs were either individually transfected (vif siRNA- 1 and vif siRNA-2) or co-transfected with 2 nM each of vif siRNA- 1 and siRNA-2 (vif siRNAl+2). (left) vif mRNA knockdown in HeLa-CD4 cells; (right) infectivity of culture supernatants from transfected HeLa-CD4 cells by TZM-bl assay. Figure 3D depicts graphs of the suppression of multiple HIV-1 strains by gag pro-siRNAs (transfected at 20 nM). Sequence of gagB siRNA (from IIIB) and its corresponding sequences in UG29 and ΓΝ22 strains were shown, (left) bar graph is gag mRNA knockdown in HeLa-CD4 cells (for IIIB), U87.CD4.CXCR4 cells (for UG29) and U87.CD4.CCR5 cells (for IN22); (right) infectivity of culture supernatants by TZM-bl assay. Data are mean±SD of 3 (Figures 3A-3C) and 2 (Figure 3D) independent experiments. mRNA expression and TZM-bl luciferase data are normalized to cells transfected with NC siRNA. Figure 3D discloses SEQ ID NOS 123-125, respectively, in order of appearance.
[0019] Figures 4A-4F demonstrating pro-siRNA sequences and tests of off-target effects. Figure 4A depicts a graph of length distribution of EGFPFL, EGFP 100 and LMNA pro-siRNAs assessed by deep sequencing. Figure 4B depicts a graph of the percentage of sequence content of all aligned deep sequencing reads. Figure 4C depicts graphs of the distribution of aligned deep sequencing reads of EGFPFL, EGFP 100 and LMNA pro-siRNAs. Figure 4D depicts volcano plots of expression changes versus p value of all annotated transcripts detected by RNA deep sequencing in HeLa-dlEGFP cells transfected with EGFP siRNAs or pro-siRNAs relative to expression in cells transfected with a negative control (NC) siRNA. Arrows indicate EGFP and the number is its fold change. Cut-off of significance is q_value<0.05 (default in Cufflinks). Figure 4E depicts volcano plots of expression changes (1.2 fold less or more) versus p value detected by microarray in HeLa-dlEGFP cells transfected with LMNA siRNAs or pro-siRNAs relative to expression in cells transfected with a NC siRNA. Arrows indicate LMNA and the number is its fold change. Cut-off of significance is p<Q.Q5 (by paired T-test). Figure 4F depicts a graph of the percentage of significantly changed transcripts in Figures 4D-4E.
[0020] Figures 5A-5B demonstrate that ectopic expression of pi 9 stabilizes -21 nt small RNA species in Listeria monocytogenes. Figure 5A depicts an image of gel demonstrating that approximately 21 nt small RNAs co-purify with pi 9. L. monocytogenes was transformed with an empty vector (pLIV-1) or with pLIV-1 encoding inducible N-terminal His-tagged pi 9 (pLIV-l-pl9- His). Duplicate cultures were grown in the presence of IPTG to induce protein expression. Samples were then lysed and incubated with Ni resin to purify the His-pl9 protein and any associated RNAs. pl9-bound RNAs were separated on a denaturing polyacrylamide gel stained with SYBR gold. M, RNA markers. Arrow indicates -21 nt small RNAs. Figure 5B depicts an image of an immunoblot with His antibody to confirm IPTG-dependent induction of His-pl9.
[0021] Figures 6A-6D demonstrate that SDS efficiently elutes GST-pl9-His-bound small RNAs but not GST-pl9-His protein. Figure 6A depicts an image of gel from an experiment in which GST- pl9-His protein, induced in E. coli with IPTG and purified by imidazole elution from Ni resin, was assayed by SDS-PAGE and Coomassie blue staining. Figure 6B depicts an image of a gel demonstrating that imidazole, but not SDS (0.5%), elutes GST-pl9-His protein from Ni beads. Coomassie blue staining of proteins eluted from Ni resin with imidazole (lane 1) or SDS (lane 2). Lane 3 shows proteins bound to the Ni resin before any elution, lane 4 is a sample of the SDS eluate and lane 5 shows proteins remaining on the beads after SDS elution (lane 5). M, protein markers. Figure 6C depicts an image of a gel demonstrating that SDS elution efficiently elutes GST-pl9-His- bound small RNAs. Ni resin was boiled before or after incubation with 0.5% SDS and bound RNAs were analyzed on a denaturing polyacrylamide gel stained with SYBR Gold. Arrow indicates -21 nt small RNAs, which were removed by SDS treatment. Figure 6D depicts a schematic summary of the effect of imidazole or SDS elution of material captured by Ni resin from E. coli expressing GST-pl9- His protein.
[0022] Figures 7A-7B demonstrate the dose response comparison of gene silencing by EGFP siRNAs and pro-siRNAs and test of antisense EGFP construct. Figure 7A depicts a graph of EGFP fluorescence in HeLa-dlEGFP cells transfected with either siRNAs or pro-siRNAs at the indicated concentrations. Data are a representative dose-response experiment. Figure 7B depicts schematics and a graph. Schemes of empty, EGFP antisense and hairpin plasmids used to produce pro-siRNAs. Bar graph is the percentage of EGFP expressing HeLa-dlEGFP cells after transfection of NC siRNA and pro-siRNAs (at 0.5 tiM). Data are mean±SD of 2 independent transfections.
[0023] Figures 8A-8B demonstrate that pro-siRNA knockdown of gene expression is independent of Dicer. Figure 8A depicts a graph of Negative control (NC) siRNA, EGFP siRNA or EGFPFL pro-siRNA co-transfected with pEGFP-Nl plasmid into HCT116 cells that contained a Dicer exon 5 deletion mutation (HCT116 Dicer ') . EGFP knockdown by siRNAs or pro-siRNAs, as measured by flow cytometry, occurred in Dicer-deficient cells. Data are representative of 3 independent experiments. Figure 8B depicts an image of a gel from experiments in which double stranded RNAs, siRNAs and pro-siRNAs were incubated or not with recombinant Dicer protein for 18 hrs at 37 °C. Resulting products were separated on a 20% polyacrylamide gel and stained with SYBR Gold.
[0024] Figure 9 depicts graphs demonstrating dose response comparison of gene silencing by pro-siRNAs and commercial siRNAs. Total RNAs were extracted from HeLa-dlEGFP cells 24 hrs after transfection. mRNA levels were normalized to negative control siRNA transfected cells. Two independent experiments were shown. Damachon siRNAs: siRNA-Dl-D4. siRNA of published sequence: siRNA-G.
[0025] Figures 10A-10B demonstrate that pro-siRNAs induce little expression of proinflammatory cytokines in primary monocyte-derived human macrophages. Figure 10A depicts a graph of the results of a qRT-PCR assay of the indicated proinflammatory cytokine gene mRNAs, 4 hr after treatment with the indicated concentrations of LPS, synthetic siRNAs, HPLC-purified pro- siRNAs or SDS-eluate. mRNA levels were normalized to levels in untreated cells. Figure 10B depicts a graph of the results of a qRT-PCR assay of the indicated proinflammatory cytokine gene, LMNA and IFIT1 mRNAs, 24 hrs after transfection with indicated siRNA and pro-siRNA (at 20 nM). PolyLC was used as positive control and mRNA levels were normalized to levels in mock transfected cells.
[0026] Figure 11 depicts length profile and distribution of deep sequencing reads aligned to the pro-siRNA target sequences.
[0027] Figures 12A-12E demonstrate a test of strand bias and validation of pro-siRNA 'hot spots' for EGFPFL pro-siRNA. Figure 12A depicts a graph of the position of DNA oligonucleotides (26-27 nt) used for probing EGFPFL small RNAs compared to position of aligned sequencing reads. The linear scale emphasizes sequencing hot spots. F, forward probe: R, reverse probe. Figure 12B depicts images of gels from experiments in which purified pro-siRNAs were denatured and incubated with the indicated DNA probes, and then the reaction mixture was analysed for the formation of DNA:RNA hybrids by native polyacrylamide gel electrophoresis and autoradiography, (top) short exposure; (middle) long exposure; (bottom) DNA oligonucleotides only, exposed for 1 hr, to show comparable labelling. Arrows indicate the DNA:RNA hybrids. Figure 12C depicts a graph of band intensities from Figure 12B which were quantified using Multi-gauge software (Fujifilm); Figure 12D depicts a graph of the ratio of sense to antisense signal for each pair of probes calculated by dividing the DNA:RNA hybrid band intensities detected with the "R" oligonucleotide by that detected with the "F" oligonucleotide. Figure 12E depicts a graph of normalized levels (to Sil) of hybridization signals (from Figure 12C) and numbers of deep sequencing reads (from Table 2) of the three hot spots.
[0028] Figures 13A-13E demonstrate the similarity of EGFPFL pro-siRNA sequence contents and hot spot patterns obtained in two independent pro-siRNA preparations. Graphs are depicted, comparing gene knockdown of EGFP measured by flow cytometry (Figure 13 A) sequence content (Figure 13B), length profile (Figure 13C) and distribution (Figure 13D) of deep sequencing reads of two independent EGFPFL pro-siRNAs {EGFPFL-1 and EGFPFL-2). Figure 13E depicts a graph comparing deep sequencing reads profiles of pro-siRNAs made from top (1-360 nt, Hotspot-1) or bottom half of EGFP (361-720 nt, Hotspot-2) with pro-siRNAs made from full length EGFP (1-720 nt, EGFPFL-1). In (13D-13E) dashed lines and * highlighted shared hotspots. NC, negative control siRNA
[0029] Figures 14A-14D demonstrate the off-target effect of siRNAs and pro-siRNAs. Figure 14D depicts Venn diagrams for significantly changed genes in HeLa-dlEGFP cells transfected with EGFP siRNAs or pro-siRNAs. Figure 14B depicts volcano plots of expression changes versus p value of all annotated lincRNA by RNA deep sequencing in HeLa-dlEGFP cells transfected with EGFP siRNAs or pro-siRNAs relative to expression in cells transfected with a negative control (NC) siRNA. Cut-off of significance is q_value<0.05 (default in Cufflinks). Figure 14C depicts graphs of the number of significantly changed lincRNAs. Figure 14D depicts Venn diagrams for significantly changed genes in HeLa-dlEGFP cells transfected with LMAN siRNAs or pro-siRNAs.
[0030] Figures 15A-15C demonstrate a two-plasmid alternate method for generating pro-siRNAs in E. coli. Figure 15A depicts a schematic of method to produce pro-siRNAs in E. coli using a two plasmid approach, where one plasmid directs pi 9 expression and the other expression of dsRNA corresponding to the target sequence. Figure 15B depicts and image of a gel from experiments in which SDS eluate of pro-siRNAs targeting EGFP, produced using this two-plasmid approach from bacteria transformed with either pRSF-GST-pl9-His or pCDF-GST-pl9-His (encoding GST-pl9-His fusion protein) in combination with L4440-EGFP plasmid (encoding T7-driven sense and antisense EGFP transcripts), were separated on a native polyacrylamide gel stained with SYBR Gold. Figure 15C depicts graphs of EGFP fluorescence measured by flow cytometry in HeLa-dlEGFP cells transfected with indicated siRNA or pro-siRNA (-10 nM). Data are representative of 3 independent experiments.
[0031] Figure 16 demonstrates an exemplary method to improve yield of pro-siRNAs. pGEX- 4T-l-pl9-T7 plasmid containing EGFP hairpin (used to make EGFPFL pro-siRNA) was co- transfected with pi 9 overexpressing plasmids (pCDF-pl9 or pRSF-pl9) or E. coli RNase III overexpressing plasmid (pCDF-RNase III). The two-plasmid system (Figures 15A-15C) of co- transfecting L4440-EGFP with pCDF-pl9 or pRSF-pl9 was also tested. All E. coli cells were cultured under the same conditions. pro-siRNAs were produced as in Figure 2A and equal proportions of SDS eluate were separated on a native polyacrylamide gel and stained with SYBR Gold. -21 nt small RNA band was quantified using Gel Logic software and signals were normalized to the band in the first sample lane. Total RNA samples extracted from E. coli cells of each condition, treated with or without RNase A, were separated on a 0.8% agarose gel containing EtBr. Immunoblots were performed to confirm expressing of pi 9 and RNase III. M, molecular weight marker. DETAILED DESCRIPTION
[0032] Embodiments of the invention described herein are directed to methods and compositions relating to the production of siRNAs in vivo, e.g. in bacterial cells (siRNAs produced according to the methods and compositions described herein are also referred to herein as "pro-siRNAs"). The technology described herein is derived from the inventors' discovery that prokaryotic cells have the ability to generate siRNAs (e.g. pro-siRNAs). As prokaryotic cells are not known to express components of the canonical RNAi machinery (e.g. Dicer), it was previously believed that prokaryotic cells were incapable of producing siRNAs.
[0033] As described in the Examples herein, when the inventors isolated pi 9 polypeptide which was expressed in a prokaryotic cell, it was found that the pi 9 polypeptide was bound to siRNAs present in the prokaryotic cell (pro-siRNAs). In the absence of the exogenous pi 9 polypeptide, these siRNAs are undetectable. These results indicated, in contrast to existing consensus in the field, that prokaryotic cells are capable of generating siRNAs, even in the absence of the canonical siRNA machinery, e.g. Dicer. The results further indicate that the endogenous siRNAs have exceptionally short half-lives which prevent their detection and/or isolation. When the inventors expressed both a pi 9 polypeptide and a dsRNA having sequence complementary to a target RNA in the prokaryotic cell, siRNAs specific for the target RNA were generated by the prokaryotic cell. The activity of these siRNAs in silencing the target RNA expressed by a eukaryotic cell is demonstrated herein.
[0034] Embodiments described herein use endogenous biological processes to generate siRNAs from dsRNA, not requiring the use of algorithms which attempt to predict efficacious siRNA sequences. Embodiments described herein also relate to populations of multiple siRNA species, wherein the population as a whole is specific for a target RNA. Such populations of siRNA species can have reduced off-target effects and greater efficacy than single RNA species.
[0035] For convenience, the meaning of some terms and phrases used in the specification, examples, and appended claims, are provided below. Unless stated otherwise, or implicit from context, the following terms and phrases include the meanings provided below. The definitions are provided to aid in describing particular embodiments, and are not intended to limit the claimed invention, because the scope of the invention is limited only by the claims. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. If there is an apparent discrepancy between the usage of a term in the art and its definition provided herein, the definition provided within the specification shall prevail.
[0036] For convenience, certain terms employed herein, in the specification, examples and appended claims are collected here.
[0037] The terms "decrease", "reduced", "reduction", or "inhibit" are all used herein to mean a decrease by a statistically significant amount. In some embodiments, the terms "reduced", "reduction", "decrease", or "inhibit" can mean a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or more or any decrease of at least 10% as compared to a reference level. In some embodiments, the terms can represent a 100%> decrease, i.e. a non-detectable level as compared to a reference level. In the context of a marker or symptom is meant a statistically significant decrease in such level. The decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is preferably down to a level accepted as within the range of normal for an individual without such disorder.
[0038] The terms "increased", "increase", "enhance", or "activate" are all used herein to mean an increase by a statically significant amount. In some embodiments, the terms "increased", "increase", "enhance", or "activate" can mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%), or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100%) increase or any increase between 10-100%) as compared to a reference level, or at least about a 2-fold, or at least about a 3 -fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
[0039] As used herein, a "subject" means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. In some embodiments, the subject is a mammal, e.g., a primate, e.g., a human. The terms, "individual," "patient" and "subject" are used interchangeably herein.
[0040] Preferably, the subject is a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but is not limited to these examples. A subject can be male or female.
[0041] A "subject in need" of treatment for a particular condition can be a subject having that condition, diagnosed as having that condition, or at risk of developing that condition.
[0042] As used herein, the term "proteins" and "polypeptides" are used interchangeably herein to designate a series of amino acid residues connected to the other by peptide bonds between the alpha- amino and carboxy groups of adjacent residues. The terms "protein", and "polypeptide" refer to a polymer of amino acids, including modified amino acids (e.g., phosphorylated, glycated, glycosylated, etc.) and amino acid analogs, regardless of its size or function. "Protein" and
"polypeptide" are often used in reference to relatively large polypeptides, whereas the term "peptide" is often used in reference to small polypeptides, but usage of these terms in the art overlaps. The terms "protein" and "polypeptide" are used interchangeably herein when referring to a gene product and fragments thereof. Thus, exemplary polypeptides or proteins include gene products, naturally occurring proteins, homologs, orthologs, paralogs, fragments and other equivalents, variants, fragments, and analogs of the foregoing.
[0043] As used herein, the term "nucleic acid" or "nucleic acid sequence" refers to any molecule, preferably a polymeric molecule, incorporating units of ribonucleic acid, deoxyribonucleic acid or an analog thereof. The nucleic acid can be either single-stranded or double-stranded. A single-stranded nucleic acid can be one strand nucleic acid of a denatured double- stranded DNA. Alternatively, it can be a single-stranded nucleic acid not derived from any double-stranded DNA. In one aspect, the template nucleic acid is DNA. In another aspect, the template is RNA. Suitable nucleic acid molecules are DNA, including genomic DNA or cDNA. Other suitable nucleic acid molecules are RNA, including mRNA.
[0044] The term "expression" refers to the cellular processes involved in producing RNA and proteins and as appropriate, secreting proteins, including where applicable, but not limited to, for example, transcription, transcript processing, translation and protein folding, modification and processing. "Expression products" include RNA transcribed from a gene, and polypeptides obtained by translation of mRNA transcribed from a gene. The term "gene" means the nucleic acid sequence which is transcribed (DNA) to RNA in vitro or in vivo when operably linked to appropriate regulatory sequences. A gene may or may not include regions preceding and following the coding region, e.g. 5' untranslated (5'UTR) or "leader" sequences and 3' UTR or "trailer" sequences, as well as intervening sequences (introns) between individual coding segments (exons).
[0045] The term "operatively linked" includes having an appropriate start signal (e.g., ATG) in front of the polynucleotide sequence to be expressed, and maintaining the correct reading frame to permit expression of the polynucleotide sequence under the control of the expression control sequence, and, optionally, production of the desired polypeptide encoded by the polynucleotide sequence. In some examples, transcription of a nucleic acid modulatory compound is under the control of a promoter sequence (or other transcriptional regulatory sequence) which controls the expression of the nucleic acid in a cell- type in which expression is intended. It will also be understood that the modulatory nucleic acid can be under the control of transcriptional regulatory sequences which are the same or which are different from those sequences which control transcription of the naturally-occurring form of a protein.
[0046] The term "isolated" or "partially purified" as used herein refers, in the case of a nucleic acid or polypeptide, to a nucleic acid or polypeptide separated from at least one other component {e.g. , nucleic acid or polypeptide) that is present with the nucleic acid or polypeptide as found in its natural source and/or that would be present with the nucleic acid or polypeptide when expressed by a cell, or secreted in the case of secreted polypeptides. A chemically synthesized nucleic acid or polypeptide or one synthesized using in vitro transcription/translation is considered "isolated."
[0047] As used herein, the term "exogenous" refers to a substance (e.g. a nucleic acid or polypeptide) present in a cell other than its native source. The term exogenous can refer to a nucleic acid or a protein (that has been introduced by a process involving the hand of man into a biological system such as a cell or organism in which it is not normally found or in which it is found in undetectable amounts. A substance can be considered exogenous if it is introduced into a cell or an ancestor of the cell that inherits the substance. In contrast, the term "endogenous" refers to a substance that is native to the biological system or cell.
[0048] As used herein, the term "complementary" or "complementary base pair" refers to A:T and G:C in DNA and A:U in RNA. Most DNA consists of sequences of nucleotide only four nitrogenous bases: base or base adenine (A), thymine (T), guanine (G), and cytosine (C). Together these bases form the genetic alphabet, and long ordered sequences of them contain, in coded form, much of the information present in genes. Most RNA also consists of sequences of only four bases. However, in RNA, thymine is replaced by uridine (U).
[0049] As used herein, "substantially complementary" refers to a first nucleotide sequence having at least 90% complementarity over the entire length of the sequence with a second nucleotide sequence, e.g. 90%> complementary, 95% complementary, 98% complementary, 99% complementary, or 100%) complementary. Two nucleotide sequences can be substantially complementary even if less than 100%) of the bases are complementary, e.g. the sequences can be mismatched at certain bases.
[0050] As used herein, the terms "gene silencing", "silencing", or "RNAi" refer to a
phenomenon where an agent for causing RNAi, such as double-stranded RNA (dsRNA) causes the specific degradation of homologous RNA, thus suppressing the expression of gene products (see Coburn, G. and Cullen, B. (2002) J. of Virology 76:9225). This process has been described in plants, invertebrates, and mammalian cells. An RNAi agent can be substantially homologous to the target RNA gene or genomic sequence, or a fragment thereof. As used in this context, the term
"homologous" is defined as being substantially identical, sufficiently complementary, or similar to the target RNA, or a fragment thereof, to effect RNA interference of the target RNA. In addition to native RNA molecules, RNAs suitable for inhibiting or interfering with the expression of a target RNA include RNA derivatives and analogs. RNAi can be caused by any type of interfering RNA, including but are not limited to, siRNA, shRNA, endogenous microRNA and artificial microRNA. In some embodiments, the RNAi molecule is a small interfering RNA (siRNA). An RNAi agent can cause a decrease in the level of a target RNA in a cell by at least about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%) or more of the target RNA level found in the cell without the presence of the gene silencing agent. In one preferred embodiment, the target RNA levels are decreased by at least about 70%, about 80%, about 90%, about 95%, about 99% or more. [0051] As used herein, the terms "treat," "treatment," "treating," or "amelioration" refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder. The term
"treating" includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder. Treatment is generally "effective" if one or more symptoms or clinical markers are reduced. Alternatively, treatment is "effective" if the progression of a disease is reduced or halted. That is, "treatment" includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment. Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable. The term "treatment" of a disease also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
[0052] As used herein, the term "pharmaceutical composition" refers to the active agent in combination with a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry. The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0053] As used herein, the term "administering" refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site. Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
[0054] The term "statistically significant" or "significantly" refers to statistical significance and generally means a two standard deviation (2SD) difference.
[0055] Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about." The term "about" when used in connection with percentages can mean ±1%.
[0056] As used herein the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the method or composition, yet open to the inclusion of unspecified elements, whether essential or not.
[0057] The term "consisting of refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment. [0058] As used herein the term "consisting essentially of refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment.
[0059] The singular terms "a," "an," and "the" include plural referents unless context clearly indicates otherwise. Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The abbreviation, "e.g." is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation "e.g." is synonymous with the term "for example."
[0060] Definitions of common terms in cell biology and molecular biology can be found in "The Merck Manual of Diagnosis and Therapy", 19th Edition, published by Merck Research Laboratories, 2006 (ISBN 0-911910-19-0); Robert S. Porter et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9). Definitions of common terms in molecular biology can also be found in Benjamin Lewin, Genes X, published by Jones & Bartlett Publishing, 2009 (ISBN-10: 0763766321); Kendrew et al. (eds.), , Molecular Biology and
Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1 - 56081-569-8) and Current Protocols in Protein Sciences 2009, Wiley Intersciences, Coligan et al., eds.
[0061] Unless otherwise stated, the present invention can be performed using standard procedures, as described, for example in Sambrook et al., Molecular Cloning: A Laboratory Manual (3 ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (2001); Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (1995); Current Protocols in Cell Biology (CPCB) (Juan S. Bonifacino et. al. ed., John Wiley and Sons, Inc.), and Culture of Animal Cells: A Manual of Basic Technique by R. Ian Freshney, Publisher: Wiley- Liss; 5th edition (2005), Animal Cell Culture Methods (Methods in Cell Biology, Vol. 57, Jennie P. Mather and David Barnes editors, Academic Press, 1st edition, 1998) which are all incorporated by reference herein in their entireties.
[0062] Other terms are defined herein within the description of the various aspects of the invention.
[0063] Embodiments of the technology described herein include methods and compositions relating to a bacterial cell comprising a siRNA-binding polypeptide and a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially complementary to at least one target RNA. siRNA-generating enzymes (e.g. RNAses) present in the bacterial cell (e.g. either endogenous or exogenous) can generate siRNA molecules from the dsRNA, which can then be bound by the siRNA- binding polypeptide. The binding of the siRNA-binding polypeptide can enable purification of the siRNA molecules from the other constituents of the bacterial cell and prevent further degradation of the siRNA to non-siRNA substituents, e.g. dsRNAs of less than 15 nucleotides in length or individual ribonucleotides.
[0064] As used herein, "double stranded RNA" or "dsRNA" refers to RNA molecules that comprise two substantially complementary strands. Double-stranded molecules include those comprising a single RNA molecule that doubles back on itself to form a two-stranded structure, e.g. a stem-loop molecule or a hairpin molecule. In some embodiments, a dsRNA can comprise nucleic acid sequences which are not substantially complementary to other sequences of the dsRNA (i.e. a single- stranded portion of the dsRNA), for example, the loop part of a stem-loop structure. The portion of the dsRNA which comprises a nucleic acid sequence substantially complementary to a target RNA should comprise, at least in part, the double-stranded portion of a dsRNA. In some embodiments, the double-stranded portion of a dsRNA comprising a nucleic acid sequence substantially complementary to at least one target RNA can be 21 nucleotides in length or greater, e.g. 21 nucleotides or greater, 22 nucleotides or greater, 23 nucleotides or greater, 24 nucleotides or greater, 25 nucleotides or greater, 50 nucleotides or greater, 100 nucleotides or greater, 200 nucleotides or greater, 500 nucleotides or greater, 1000 nucleotides or greater in length.
[0065] As used herein, the term "target RNA" refers to a RNA present in a cell (i.e. the "target cell"). The target RNA comprises a target sequence to which one strand of a siRNA according to the methods and compositions described herein binds, thereby causing RNAi silencing of the target RNA. The target cell can be the bacterial cell comprising a siRNA-binding polypeptide or another cell, either prokaryotic or eukaryotic. The target sequence can be an RNA that can be translated (i.e. it can encode a polypeptide, e.g. mRNA) or it can be an RNA that is not translated (i.e. a non-coding RNA). In some embodiments, the target sequence can be any portion of an mRNA. In some embodiments, the target sequence can be a sequence endogenous to the cell. In some embodiments, the target sequence can be a sequence exogenous to the cell. In some embodiments, the target sequence can be sequence from an organism that is pathogenic to the target cell, e.g. the target sequence can be sequence from a viral, bacterial, fungal, and/or parasitic origin. In some embodiments, the target sequence is a viral nucleotide sequence.
[0066] In some embodiments, a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence substantially complementary to part or all of the sequence of the target RNA. In some embodiments, a dsRNA comprising a nucleic acid sequence
substantially complementary to a target RNA can comprise sequence complementary to part or all of a specific allele, variant, and/or mutation (e.g., insertions, deletions, fusions, SNPs, etc.) of a target RNA. In some embodiments, the dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise nucleic acid sequence(s) substantially complementary to multiple target RNAs (e.g. target RNAs encoding separate genes or target RNAs encoding multiple variants of the same gene). In some embodiments, a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence substantially complementary to all or part of one or more exons of a target mRNA. In some embodiments, a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence substantially complementary to a cDNA. In some embodiments, a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can comprise a nucleic acid sequence (or its complement) obtained from the transcriptome and/or genome of a cell.
[0067] In some embodiments, the dsRNA can comprise two separate complementary strands, e.g. a sense and antisense strand.
[0068] In some embodiments, the dsRNA can be a hairpin RNA, i.e. an RNA comprising two portions which are reverse complements, separated by a sequence which will not self-anneal, thus forming a stem-loop or "hairpin" structure. In some embodiments, the double-stranded portion of a hairpin RNA can be at least 19 nucleotides in length. In some embodiments, the double-stranded portion of a hairpin RNA can be at least 25 nucleotides in length. In some embodiments, the double- stranded portion of a hairpin RNA can be 30 nucleotides in length or greater, e.g. at least 30 nucleotides, at least 50 nucleotides, at least 100 nucleotides, at least 200 nucleotides, or at least 300 nucleotides. In some embodiments, the dsRNA can be a shRNA. As used herein "shRNA" or "small hairpin RNA" (also called stem loop) is a type of dsRNA. In one embodiment, these shRNAs are composed of a short, e.g. about 19 to about 25 nucleotide, antisense strand, followed by a nucleotide loop of about 5 to about 9 nucleotides, and the analogous sense strand. Alternatively, the sense strand can precede the nucleotide loop structure and the antisense strand can follow.
[0069] In some embodiments, increased length of the double-stranded portion of a dsRNA can correlate with a decreased level of off-target effects, e.g. silencing of non-targeted genes. In some embodiments, one strand of the double-stranded portion of a dsRNA can be at least 100 nucleotides in length. For example, one strand of the double-stranded portion of a dsRNA can be at least 100 nucleotides in length, at least 200 nucleotides in length, at least 300 nucleotides in length, at least 400 nucleotides in length, at least 500 nucleotides in length, at least 700 nucleotides in length, or at least 1000 nucleotides in length.
[0070] In some embodiments, the dsRNA can be exogenous to the cell. In some embodiments, the target sequence of the target RNA can be exogenous to the cell. In some embodiments, the target RNA can be exogenous to the cell. In some embodiments, the nucleic acid sequence substantially complementary to a target RNA can be exogenous to the cell.
[0071] In the methods and compositions described herein, siRNAs can be generated from the dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. As used herein, the term "siRNA" refers to a nucleic acid that forms an RNA molecule comprising two individual strands of RNA which are substantially complementary to each other. Typically, the siRNA is at least about 15-40 nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is about 15-40 nucleotides in length, and the double stranded siRNA is about 15-40 base pairs in length, preferably about 19-25 base nucleotides, e.g., 19, 20, 21, 22, 23, 24, or 25 nucleotides in length). In some embodiments, a siRNA can be blunt-ended. In some embodiments, a siRNA can comprise a 3' and/or 5' overhang on each strand having a length of about 0, 1, 2, 3, 4, or 5 nucleotides. The length of the overhang is independent between the two strands, i.e., the length of the overhang on one strand is not dependent on the length of the overhang on the second strand. The siRNA molecules can also comprise a 3 ' hydroxyl group. In some embodiments, the siRNA can comprise a 5' phosphate group. A siRNA has the ability to reduce or inhibit expression of a gene or target RNA when the siRNA is present or expressed in the same cell as the target gene, e.g. the target RNA. siRNA-dependent post-transcriptional silencing of gene expression involves cutting the target RNA molecule at a site guided by the siRNA.
[0072] In some embodiments, a single siRNA species can be generated from a dsRNA. In some embodiments, multiple siRNA species can be generated from a dsRNA. For example, two or more siRNA species can be generated from a dsRNA, e.g. two or more siRNA species, three or more siRNA species, five or more siRNA species, or ten or more siRNA species. As used herein, the term "a siRNA species" refers to one or more siRNA molecules which are identical in sequence. In embodiments where multiple siRNA species are generated from a single dsRNA, the species can comprise sequence complementary to the same target RNA or to separate target RNAs. In some embodiments, a single dsRNA can comprise sequence complementary to multiple target RNAs. In some embodiments, a single dsRNA can comprise multiple sequences, each of which is
complementary to a unique target RNA, e.g. a multiplicity of siRNA species targeting (e.g.
complementary to) a multiplicity of target RNAs can be generated from a single dsRNA.
[0073] In the methods and compositions described herein, a dsRNA present within a bacterial cell can be cleaved to generate one or more siRNA species. The siRNA molecules can then be bound by a siRNA-binding polypeptide also present within the bacterial cell. As used herein, the term "siRNA-binding polypeptide" refers to a polypeptide capable of binding to siRNAs and increasing the half-life or detectable level of siRNAs in a prokaryotic cell. In some embodiments, the siRNA- binding polypeptide can bind preferentially or specifically to siRNAs as compared to other dsRNA species, e.g. the polypeptide can bind preferentially or specifically to siRNAs as compared to dsRNAs greater than 25 or less than 15 nucleotides in size. In some embodiments, the siRNA-binding polypeptide can bind preferentially or specifically to siRNAs as compared to other dsRNA species, e.g. dsRNAs greater than 25 or less than 15 nucleotides in size. In one embodiment, the siRNA- binding polypeptide does not bind to dsRNA having a double-stranded portion longer than 25 nucleotides in length. In some embodiments, the siRNA-binding polypeptide can bind preferentially or specifically to siRNAs as compared to single-stranded RNA species.
[0074] In some embodiments, a siRNA-binding polypeptide can detectably bind to a siRNA. In some embodiments, a siRNA-binding polypeptide can be a polypeptide that when expressed in a bacterial cell, can cause detectable levels of siRNAs to be present in that cell when detectable levels of siRNAs are not present in the wild-type bacterial cell. In some embodiments, a siRNA-binding polypeptide can be a polypeptide that increases the half-life or detectable level of siRNAs in a prokaryotic cell by at least 5%, e.g. by at least 5%, by at least 10%, by at least 20%, by at least 30%, by at least 50%, by at least 75%, by at least 100%, by at least 200% or more.
[0075] In some embodiments, a siRNA-binding polypeptide can be a pi 9 polypeptide. As used herein, the term "pi 9" refers to a viral protein which binds specifically to dsRNAs and which suppresses RNAi-mediated host plant viral defenses. The sequences of pi 9 polypeptides from a number of species are known, e.g. tombusvirus pl9 (NCBI Gene ID: 1493957; SEQ ID NO: l). In some embodiments, the pl9 polypeptide can be tombusvirus pl9. Non-limiting examples of pl9 homologues include Carnation Italian ringspot virus PI 9; Tomato bushy stunt virus pi 9; Artichoke mottled crinkle virus pl9; Lisianthus necrosis virus pl9; Pear latent virus pl9; Cucumber Bulgarian virus pl9; Cucumber necrosis virus pl9; Pelargonium necrotic spot virus pl9; Cymbidium ringspot virus pl9; Lisianthus necrosis virus pl9; Lettuce necrotic stunt virus pl9; Maize necrotic streak virus pl9; Grapevine Algerian necrosis virus pl9; and Grapevine Algerian latent virus pl9. A pl9 polypeptide can comprise mutants, variants, homologues, and functional fragments of wildtype pi 9 polypeptides.
[0076] Further non-limiting examples of an siRNA-binding polypeptide can include the Flock house virus B2; HC-Pro; Tobacco etch virus HC-Pro; P38; P122; P130; Tobamovirus P122/P130; p21 ; Rice hoja blanca tenuivirus (RHBV) NS3; Cucumber vein yellowing virus Plb; HC-Pro of potyviruses; p21 of Beet yellows virus and Closterovirus; and variants, homologues, or functional fragments of the foregoing.
[0077] In some embodiments, an siRNA-binding polypeptide can be an enzymatically inactive member of the RISC complex, e.g. an enzymatically inactive variant or mutant of Argonaute or Dicer (see, e.g. Buker et al. Nat Struct Mol Bio 2007 14:200-7 and Liu et al. Molecular Cell 2012 46: 1-11 ; which are incorporated by reference herein in their entireties). In some embodiments, the siRNA- binding polypeptide is not an enzymatically active member of the RISC complex, e.g. an Argonaute or RISC polypeptide. As used herein, the term "RISC complex" refers to the proteins and single- stranded polynucleotides that interact to recognize target RNA molecules. Demonstrated components of RISC include the Argonaute proteins (e.g. Aubergine, Argonaute 2), R2D2, and Dicer (e.g. Dcr-2). In the case of an active RISC complex loaded with a single-stranded guide RNA derived from a siRNA, the RISC complex can cleave the target RNA molecule.
[0078] In some embodiments, a siRNA-binding polypeptide can be a polypeptide that can bind to nucleic acids, e.g. protamine, or a variant, homologue, or functional fragment thereof (see, e.g. Rossi. Nature Biotechnology 2005 23:682-4 and Reischl et al. Scientia Pharmaceutica 2010 78:686; which are incorporated by reference herein in their entirety). In some embodiments, a siRNA-binding polypeptide can be a polypeptide that can be bind to dsRNAs, e.g. TARBP2 or a polypeptide comprising a double-stranded RNA binding domain (see, e.g. US Patent Publication 2009/0093026; which is incorporated by reference herein in its entirety) or a variant, homologue, or functional fragment thereof.
[0079] A functional fragment of a siRNA-binding polypeptide can be any portion of a siRNA- binding polypeptide which retains at least 50% of the wild-type level of siRNA binding activity, e.g. at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or more.
[0080] In some embodiments, a siRNA-binding polypeptide can comprise a purification tag. The term "purification tag" as used herein refers to any peptide sequence suitable for purification of a siRNA-binding polypeptide, and optionally, siRNAs bound by the siRNA-binding polypeptide. The purification tag specifically binds to (or is bound by) another moiety with affinity for the purification tag. Such moieties which specifically bind to a purification tag can be attached to a matrix or a resin, e.g. agarose beads. Moieties which specifically bind to purification tags can include antibodies, nickel or cobalt ions or resins, biotin, amylose, maltose, and cyclodextrin.
Exemplary purification tags can include histidine tags (such as a hexahistidine peptide (SEQ ID NO: 122)), which will bind to metal ions such as nickel or cobalt ions. Therefore, in certain embodiments the purification tag can comprise a peptide sequence which specifically binds metal ions. Other exemplary purification tags are the myc tag (EQKLISEEDL (SEQ ID NO:3)), the Strep tag
(WSHPQFEK (SEQ ID NO:4)), the Flag tag (DYKDDDDK (SEQ ID NO:5)) and the V5 tag (GKPIPNPLLGLD ST (SEQ ID NO:6)). The term "purification tag" also includes "epitope tags", i.e. peptide sequences which are specifically recognized by antibodies. Exemplary epitope tags can include the FLAG tag, which is specifically recognized by a monoclonal anti-FLAG antibody. The peptide sequence recognized by the anti-FLAG antibody consists of the sequence DYKDDDDK (SEQ ID NO: 5) or a substantially identical variant thereof. Therefore, in certain embodiments the purification tag can comprise a peptide sequence which is specifically recognized by an antibody. The term "purification tag" also includes substantially identical variants of purification
tags. "Substantially identical variant" as used herein refers to derivatives or fragments of purification tags which are modified compared to the original purification tag (e.g. via amino acid substitutions, deletions or insertions), but which retain the property of the purification tag of specifically binding to a moiety which specifically recognizes the purification tag. In some embodiments, the siRNA- binding polypeptide can be a pi 9 fusion protein as described in US Patent Publication 2010/0209933; which is incorporated herein by reference in its entirety.
[0081] In some embodiments, the siRNA-binding polypeptide can be encoded by a nucleic acid present in the bacterial cell, i.e. the polypeptide is transcribed and translated by the bacterial cell. In some embodiments, the siRNA-binding polypeptide can be introduced into the bacterial cell as a polypeptide. Uptake of polypeptides can be induced by any means in the art. Non-limiting examples include the protocols described in Shellman and Pettijohn. J Bacteriology 1991 173:3047-3059; which is incorporated by reference herein in its entirety. [0082] In some embodiments, a nucleic acid encoding a siRNA-binding polypeptide and/or a nucleic acid encoding a dsRNA can be present within the bacterial genome, e.g. the nucleic acids can be incorporated into the genome. In some embodiments, a nucleic acid encoding a siRNA-binding polypeptide and/or a nucleic acid encoding a dsRNA can be present within a vector. In some embodiments, a nucleic acid encoding a siRNA-binding polypeptide and/or a nucleic acid encoding a dsRNA can be present within portions of the same vector. In some embodiments, the nucleic acids encoding the siRNA-binding polypeptide and the dsRNA can be present within portions of separate vectors.
[0083] The term "vector", as used herein, refers to a nucleic acid construct designed for delivery to a host cell or transfer between different host cells. As used herein, a vector can be viral or non- viral. Many vectors useful for transferring exogenous genes into target cells are available, e.g. the vectors may be episomal, e.g., plasmids, virus derived vectors or may be integrated into the target cell genome, through homologous recombination or random integration. In some embodiments, a vector can be an expression vector. As used herein, the term "expression vector" refers to a vector that has the ability to incorporate and express heterologous nucleic acid fragments in a cell. An expression vector may comprise additional elements, for example, the expression vector may have two replication systems, thus allowing it to be maintained in two organisms. The nucleic acid incorporated into the vector can be operatively linked to an expression control sequence when the expression control sequence controls and regulates the transcription and translation of that polynucleotide sequence. In some embodiments, the dsRNA and the nucleic acid encoding the siRNA-binding polypeptide can be within the same operon. In some embodiments, the dsRNA and the nucleic acid encoding the siRNA-binding polypeptide can be within separate operons.
[0084] In some embodiments, a siRNA-binding polypeptide and/or dsRNA encoded by a nucleic acid can be present within a portion of a plasmid. Plasmid vectors can include, but are not limited to, pBR322, pBR325, pACYC177, pACYC184, pUC8, pUC9, pUC18, pUC19, pLG339, pR290, pKC37, pKClOl, SV 40, pBluescript II SK +/- or KS +/- (see "Stratagene Cloning Systems" Catalog (1993) from Stratagene, La Jolla, Calif, which is hereby incorporated by reference), pQE, pIH821, pGEX, pET series (see Studier et. al., "Use of T7 RNA Polymerase to Direct Expression of Cloned Genes," Gene Expression Technology, vol. 185 (1990), which is hereby incorporated by reference in its entirety).
[0085] As used herein, the term "viral vector" refers to a nucleic acid vector construct that includes at least one element of viral origin and has the capacity to be packaged into a viral vector particle. The viral vector can contain a transgenic gene in place of non-essential viral genes. The vector and/or particle may be utilized for the purpose of transferring any nucleic acids into cells either in vitro or in vivo. Numerous viral vectors are known in the art and can be used as carriers of a nucleic acid into a cell, e.g. lambda vector system gtl 1, gt WES.tB, Charon 4. [0086] In accordance with the methods and compositions described herein, siRNAs specific for the target RNA can be produced in a bacterial cell when both the dsRNA and the siRNA-binding polypeptide are present and/or expressed. In some embodiments, the dsRNA and/or the siRNA- binding polypeptide can be constitutively expressed. In some embodiments, nucleic acids encoding the dsRNA and/or the siRNA-binding polypeptide can be operably linked to a constitutive promoter. In some embodiments, the dsRNA and/or the siRNA-binding polypeptide can be inducibly expressed. In some embodiments, nucleic acids encoding the dsRNA and/or the siRNA-binding polypeptide can be operably linked to an inducible promoter.
[0087] As described herein, an "inducible promoter" is one that is characterized by initiating or enhancing transcriptional activity when in the presence of, influenced by, or contacted by an inducer or inducing agent than when not in the presence of, under the influence of, or in contact with the inducer or inducing agent. An "inducer" or "inducing agent" may be endogenous, or a normally exogenous compound or protein that is administered in such a way as to be active in inducing transcriptional activity from the inducible promoter. In some embodiments, the inducer or inducing agent, e.g., a chemical, a compound or a protein, can itself be the result of transcription or expression of a nucleic acid sequence ( e.g., an inducer can be a transcriptional repressor protein), which itself may be under the control or an inducible promoter. Non-limiting examples of inducible promoters include but are not limited to, the lac operon promoter, a nitrogen-sensitive promoter, an IPTG- inducible promoter, a salt-inducible promoter, and tetracycline, steroid-responsive promoters, rapamycin responsive promoters and the like. Inducible promoters for use in prokaryotic systems are well known in the art, see, e.g. the beta. -lactamase and lactose promoter systems (Chang et al., Nature, 275: 615 ( 1978, which is incorporated herein by reference); Goeddel et al.. Nature, 281 : 544 (1979), which is incorporated ereirs by reference), the arahinose promoter system, including the araBAD promoter (Guzman et al. J . Bacteriol., 174: 7716-7728 (1 992), which is incorporated herein by reference; Guzman et al., J. Bacteriol., 177: 4.121-4130 (1995), which is incorporated herein by reference; Siegele and Flu. Proc. Natl. Acad. Sci. USA, 94: 8168-8172 (1997), which is incorporated herein by reference), the rhamnose promoter (Haldimann et al, J. Bacteriol., 180: 1277-1286 (1998), which is incorporated herei by reference), the alkaline phosphatase promoter, a tryptophan (trp) promoter system (Goeddel, Nucleic Acids Res., 8: 4057 (1980), which is incorporated herein by reference), the P;.¾to- ; and Ρ^^ι promoters (Lutz and Bujard, Nucleic Acids Res., 25: 1203-1210 (1997), which is incorporated herein by reference), and hybrid promoters such as the tac promoter. deBoer et ah, Proc. Natl. Acad. Sci. USA, 80: 21 -25 (1983), which is incorporated herein by reference.
[0088] An inducible promoter useful in the methods and systems as disclosed herein can be induced by one or more physiological conditions, such as changes in pH, temperature, radiation, osmotic pressure, saline gradients, cell surface binding, and the concentration of one or more extrinsic or intrinsic inducing agents. The extrinsic inducer or inducing agent may comprise amino acids and amino acid analogs, saccharides and polysaccharides, nucleic acids, protein transcriptional activators and repressors, cytokines, toxins, petroleum-based compounds, metal containing compounds, salts, ions, enzyme substrate analogs, hormones, and combinations thereof. In specific embodiments, the inducible promoter is activated or repressed in response to a change of an environmental condition, such as the change in concentration of a chemical, metal, temperature, radiation, nutrient or change in pH. Thus, an inducible promoter useful in the methods and systems as disclosed herein can be a phage inducible promoter, nutrient inducible promoter, temperature inducible promoter, radiation inducible promoter, metal inducible promoter, hormone inducible promoter, steroid inducible promoter, and/or hybrids and combinations thereof. Appropriate environmental inducers can include, but are not limited to, exposure to heat (i.e., thermal pulses or constant heat exposure), various steroidal compounds, divalent cations (including Cu2+ and Zn2+), galactose, tetracycline, IPTG (isopropyl-β- D thiogalactoside), as well as other naturally occurring and synthetic inducing agents and gratuitous inducers.
[0089] Inducible promoters useful in the methods and systems as disclosed herein also include those that are repressed by "transcriptional repressors" that are subject to inactivation by the action of environmental, external agents, or the product of another gene. Such inducible promoters may also be termed "repressible promoters" where it is required to distinguish between other types of promoters in a given module or component of the biological switch converters described herein. Preferred repressors for use in the present invention are sensitive to inactivation by physiologically benign agent. Thus, where a lac repressor protein is used to control the expression of a promoter sequence that has been engineered to contain a lacO operator sequence, treatment of the host cell with IPTG will cause the dissociation of the lac repressor from the engineered promoter containing a lacO operator sequence and allow transcription to occur. Similarly, where a tet repressor is used to control the expression of a promoter sequence that has been engineered to contain a tetO operator sequence, treatment of the host cell with tetracycline will cause the dissociation of the tet repressor from the engineered promoter and allow transcription of the sequence downstream of the engineered promoter to occur.
[0090] A bacterial cell of the methods and compositions described herein can be any of any species. Preferably, the bacterial cells are of a species and/or strain which is amenable to culture and genetic manipulation. In some embodiments, the bacterial cell can be a gram-positive bacterial cell. In some embodiments, the bacterial cell can be a gram-negative bacterial cell. In some embodiments, the parental strain of the bacterial cell of the technology described herein can be a strain optimized for protein expression. Non-limiting examples of bacterial species and strains suitable for use in the present technologies include Escherichia coli, E. coli BL21, E. coli Tuner, E. coli Rosetta, E. coli JM101, and derivatives of any of the foregoing. Bacterial strains for protein expression are commercially available, e.g. EXPRESS™ Competent E. coli (Cat. No. C2523; New England Biosciences; Ipswich, MA). [0091] A dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA can be processed to create siRNA molecules by a siRNA-generating enzyme (e.g. RNAse III) present within the bacterial cell. In some embodiments, the bacterial cell can be a cell which expresses a siRNA-generating polypeptide. In some embodiments, the bacterial cell can be a cell which overexpresses a siRNA-generating polypeptide. As used herein, a "siRNA-generating polypeptide" refers to an enzyme with RNase activity which can cleave dsRNA in such a way that siRNAs result. In some embodiments, the siRNA-generating polypeptide can be an RNaselll polypeptide. As used herein the term "RNaselll polypeptide" refers to a eukaryotic class I RNase III, e.g. E. coli RNaselll (NCBI Gene ID: 947033; SEQ ID NO: 2). siRNA-generating polypeptides can be mutants, variants, homologues, or functional fragments of wildtype siRNA-generating polypeptides which retain at least 50% of the siRNA generating activity of the wildtype, e.g. at least 50%, at least 60%>, at least 70%, at least 80%), at least 90%> or more of the wildtype activity. In some embodiments, the siRNA- generating enzyme can be endogenous to the bacterial cell. In some embodiments, the siRNA- generating enzyme can be exogenous to the bacterial cell.
[0092] In some embodiments, a cell can comprise a mutation and/or transgene which enhances the expression and/or activity of a siRNA-generating polypeptide. By way of non- limiting example, a cell can comprise a mutation in the endogenous RNaselll promoter which increases expression, or a cell can comprise a transgenic (e.g. exogenous) construct with an RNaselll gene under the control of a strong constitutive or inducible promoter, or a cell can comprise a nucleic acid encoding a polypeptide which increases the activity and/or expression of RNaselll, e.g. the T4 polynucleotide
kinase/phosphatase (PNK) (see Durand et al. PNAS 2012 109:7073-8; which is incorporated by reference herein in its entirety). In some embodiments, a cell can express an ectopic level and/or amount of a siRNA-generating polypeptide (e.g. RNaselll). As used herein, "ectopic" refers to a substance that is found in an unusual location and/or amount. An ectopic substance can be one that is normally found in a given cell, but at a lower amount and/or at a different time.
[0093] In one aspect, the technology described herein relates to a method of producing one or more siRNA species which can inhibit the expression of a target RNA, the method comprising culturing a bacterial cell comprising at least a siRNA-binding polypeptide and a dsRNA wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA under conditions suitable for the production of siRNAs. As used herein, the term "conditions suitable for the production of siRNAs" refers to conditions under which a siRNA-generating enzyme within a bacterial cell cleaves the dsRNA in the presence of a siRNA-binding polypeptide. In embodiments wherein one or more of the dsRNA and the siRNA-binding polypeptide are encoded by nucleic acids, conditions suitable for the production of siRNAs can include conditions under which the cell will express (i.e. transcribe and, in some cases, translate) the dsRNA and/or the siRNA-binding polypeptide from the nucleic acid. The precise conditions will vary depending on the exact identity of the bacterial cell, the presence of other exogenous DNA or mutations, and whether or not a nucleic acid encoding a dsRNA and/or siRNA-binding polypeptide is operably linked to an inducible or constitutive promoter. In some embodiments, wherein the nucleic acid(s) encoding a dsRNA and/or siRNA-binding polypeptide are operably linked to inducible promoters, conditions suitable for the production of siRNAs can include conditions which induce expression from the inducible promoter, e.g. permissive temperatures and/or the presence of compounds which induce expression from the inducible promoter. In some embodiments, conditions suitable for the production of siRNAs can include conditions which encourage exponential growth of the bacterial cells. By way of non-limiting example conditions suitable for the production of siRNAs in E. coli T7 Express Iq (NEB) can include LB broth, Lennox (BD) at 37°C with shaking at 250 rpm and appropriate antibiotics.
[0094] In some embodiments, a method of producing one or more siRNA species can further comprise isolating the siRNA-binding polypeptide and eluting the siRNAs bound to the siRNA- binding polypeptide. In some embodiments, the siRNA-binding polypeptide can be isolated via a purification tag as described elsewhere herein.
[0095] In some embodiments, the siRNAs bound to a siRNA-binding polypeptide can be eluted from the isolated siRNA-binding polypeptide. Methods of eluting nucleic acids from proteins are well known in the art. By way of non-limiting example, siRNAs can be eluted from a siRNA-binding polypeptide by contacting the polypeptide-siRNA complex with a solution comprising 0.5% SDS for 10 min at room temperature with rotation. The solution can then be collected and passed through a 0.22 μηι centrifuge filter (Corning).
[0096] In some embodiments, the siRNAs eluted from a siRNA-binding polypeptide can be further purified. Methods of nucleic acid purification are well known in the art and include, but are not limited to anion exchange HPLC, PAGE purification, desalting, and filtration. See, e.g. Gjerde et al. "RNA Purification and Analysis" Wiley-VCH; 2009 and Farrell et al. "RNA Methodologies" 4th Ed., Academic Press; 2010. In some embodiments, the siRNAs eluted from a siRNA-binding polypeptide can be further purified by HPLC.
[0097] In some embodiments, the siRNAs can be isolated from the totality of the cell contents without first isolating the siRNAs bound to siRNA-binding polypeptides. Methods of purifying RNA molecules are well known in the art, as described above, and any method or combination of methods known in the art can be used to isolate and/or purify the siRNAs produced according to the methods described herein.
[0098] In some embodiments, the methods described herein can further comprise contacting the bacterial cell with one or more modified nucleotides before or during the culturing step, thereby causing one or more modified nucleotides to be incorporated into the siRNA(s) of the presently described technologies. A modified nucleotide can be any nucleotide other than adenine "A", guanine "G", uracil "U", or cytosine "C". Such modified nucleotides include nucleotides which contains a modified sugar moiety, a modified phosphate moiety ami/or a modified nucleobase. A modified nucleotide residue or a derivative or analog of a natural nucleotide are also useful. Examples of modified residues, derivatives or analogues include, but are not limited to, aminoallyl UTP, pseudo- UTP, 5-I-UTP, 5-I-CTP, 5-Br-UTP, alpha-S ATP, alpha-S CTP, alpha-S GTP, alpha-S UTP, 4-thio UTP, 2-thio-CTP, 2TSTH2 UTP, 2TSTH2 CTP, and 2'F UTP. Such modified nucleotides include, but are not limited to, aminoallyl uridine, pseudo-uridine, 5-I-uridine, 5-I-cytidine, 5-Br-uridine, alpha-S adenosine, alpha-S cytidine, alpha-S guanosine, alpha-S uridine, 4-thio uridine, 2-thio-cytidine, 2TSTH2 uridine, 2ΉΉ2 cytidine, and 2' F uridine, including the free pho (NTP) RNA molecules as well as all other useful forms of the nucleotides. Further non-limiting examples of modified nucleotides can include ribonucleotides having a 2'-0-metbyl (TOMe), 2i-deoxy-2,fluoro, 2'-deoxy, 5-C-methyl, 2'-methoxyethyl, 4'-thio, 2'-amino, or 2'-C-allyl group, locked nucleic acid (LNA) nucleotides (e.g.,
2 - 0, 4'-C -methylene -(D -ribofuranosyl) nucleotides), 2'-methoxyethoxy (MOE) nucleotides, 2'~ methylthio-ethyl nucleotides, 2'-deoxy-2'-fluoro nucleotides, 2'-deoxy-2'-cb.loro nucleotides, and 2 - azido nucleotides. ), nucleotides having a nucleotide base analog such as. for example, C-phenyl, C- naphthyl, other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as
3 - nitropyrrole, 4-mtfoindole, S -nitroindole, and 6-nitroindoIe. Modification of the sugar moiety can include, but is not limited to, replacement of the ribose ring with a hexose, cyelopentyl or cyclobexyl ring. Alternatively, the D-ribose ring of a naturally-occurring nucleic acid can be replaced with an L- ribose ring or the (3-atiomer of a naturally occurring nucleic acid can be replaced with the a-anomer. Modified phosphate moieties can include phosphorothioates, phosphorodithioates, methyl phosphorates, alkylphospbonates, alkylphosphonothioates, methyl phosphates, phosphoramidat.es, and the like, or combinations thereof. Oligonucleotides which comprise such modified phosphate linkages can have impro ved properties when compared to corresponding oligonucleotides comprising only phosphate diester linkages, e.g. increased resistance to degradation by nucleases. Modified nucieobases include 7-deazaguanine, 7-deaza-8-azaguanine, 5-propyny!cytosine, 5-propynyiuricil, 7- deazaadenine. 7-deaza-8azaadenine, 7-deaza-6-oxopurine, 6-oxopurine, 3-deazaadenosine, 2-oxo-5- methylpyrhnidine, 2-oxo-4-methylthio-5methylp<yrimidine, 2-thiocarbonyl-4-oxo-5methylpyrimidine,
4- oxo-5-methylpyrimidine, 2-ammopurme, 5-fluoronricil, 2,6-diaminopurine, 8-aminopurine, 4triazoio · 5-methylthyniine, and 4 -triazolo- 5 -mefhyluricii. Modified nucieobases can also include abasic moieties. Additional non-limiting examples of modified nucleotides include biotinylated nucleotides, amine -modifi d nucleotides, alkylated nucleotides, fluorophore-labeled nucleotides, radiolabeled nucleotides, phosphorothioates, phosphoramidites, phosphites, ring atommodified derivatives and the like. In some embodiments, a modified nucleotide can be a G-clamp nucleotide. A G-clamp nucleotide refers to a modified cytosine analog wherein the modifications confer the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine nucleotide within a duplex (see, e.g., Lin et al., J Am. Chem. Soc, 120:8531-8532 ( 998); which is incorporated herein by reference in its entirety, in some embodiments, a modified nucleotide can comprise multiple modifications, ϊη some embodiments, a cell can be contacted with any combination of modified nucleotides. [0099] In one aspect, the technology described herein relates to a library of siRNA species, the library comprising a plurality of clonal bacterial cell populations; wherein each clonal population comprises bacterial cells as described herein. In some embodiments, the bacterial cells can comprise at least a siRNA-binding polypeptide and a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA. In some embodiments, wherein a bacterial cell comprises dsRNAs specific for a single target RNA, the clonal bacterial population comprising that cell can comprise a population of siRNAs which will specifically bind to the single target RNA and/or which will specifically silence the expression of the target RNA.
[00100] In one aspect, the technology described herein relates to a library of siRNA species, the library comprising a plurality of populations of siRNAs; wherein each population of siRNAs is obtained according to the methods described herein. As used herein, a "population of siRNAs" refers to two or more siRNAs, wherein at least two of the siRNAs comprise non-identical sequences, but wherein the two or more siRNAs each comprise a nucleic acid sequence substantially complementary to the same target RNA. For example, a population of siRNAs can comprise two more siRNA species. In some embodiments, a population of siRNAs can be generated from a single dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA. In some embodiments, a population of siRNAs can bind to a single target RNA and/or can specifically silence the expression of the target RNA. A population of siRNAs can be present within a bacterial cell or isolated from a bacterial cell.
[00101] Methods of creating bacterial libraries, and/or libraries of compounds isolated from bacterial cells are well known in the art. By way of non-limiting example, a bacterial cell library can be in the form of a plurality of multi-well plates, with each well of a plate comprising a clonal bacterial population. The clonal bacterial populations can be provided in media or in glycerol stocks. In some embodiments, a library can comprise multiple wells which comprise identical clonal populations, i.e. a clonal population can appear multiple times in a library. In some embodiments, a library can comprise a plurality of multi-well plates, with each well of a plate comprising one or more siRNA species (e.g. a siRNA species or a population of siRNA species) isolated from one or more clonal bacterial populations. Methods of isolating nucleic acids from bacterial cells are well known in the art and examples are described elsewhere herein. In some embodiments, libraries can be created using automated and/or high-throughput methods, e.g. robotic colony-picking.
[00102] In some embodiments, a library can comprise pooled samples, e.g. multiple clonal bacterial populations, multiple isolated siRNAs, or multiple isolated populations of siRNA species can be pooled so that a smaller number of samples must be initially screened. The individual components of a "positive" pooled can be subsequently screened separately.
[00103] In some embodiments, a library can comprise 10 or more pools of, populations of, and/or individual siRNA species (e.g. isolated or present within bacterial cells), e.g. 10 or more, 100 or more, 1,000 or more, 10,000 or more, or 100,000 or more pools of, populations of, and/or individual siRNA species.
[00104] In some embodiments, a library can comprise a plurality of populations of siRNAs, wherein each population of siRNAs can silence at least one target RNA of a target set. A target set of RNAs can comprise, e.g. the transcriptome of a cell, the transcriptome of an organism, the transcriptome of a cell and/or organism in a specific state (e.g. a diseased organism or an organism at a specific stage of development) or a subtractive transcriptome (e.g. all the transcripts present in a cell under one condition but which are not present in the cell in a second condition).
[00105] In one aspect, the technology described herein relates to vectors which enable the use of the methods and compositions described herein. In some embodiments, the vector can be an expression vector. In some embodiments, the vector can be a plasmid. In some embodiments, a vector for use in the methods and compositions described herein can comprise (a) a nucleic acid encoding a siRNA-binding polypeptide and (b) a dsRNA cloning site. In some embodiments, a dsRNA cloning site further can further comprise a nucleic acid encoding a dsRNA, wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA. As used herein, a "dsRNA cloning site" refers to a multiple cloning site comprising at least one restriction enzyme site and which can accept the insertion of nucleic acid sequence(s) comprising the sequence of both a sense and anti-sense strand of nucleic acid; wherein one strand is substantially
complementary to the nucleic acid sequence of a target RNA, such that a dsRNA will be encoded and can be expressed, e.g. a sequence inserted at the dsRNA cloning site will be operably linked to a promoter as described herein. In some embodiments, a single nucleic acid molecule can comprise the sequence of both the sense and anti-sense strand prior to insertion at the dsRNA cloning site. In some embodiments, a dsRNA cloning site can comprise a nucleic acid sequence which comprises sequences which can be cleaved by at least two different restriction enzymes.
[00106] In some embodiments, a dsRNA cloning site can comprise a nucleic acid sequence which comprises sequences which can be cleaved by at least four different restriction enzymes. In some embodiments, a dsRNA cloning site can comprise two multiple cloning sites separated by a nucleic acid sequence encoding a hairpin sequence; wherein each multiple cloning site comprises a nucleic acid sequence which comprises sequences which can be cleaved by at least two different restriction enzymes. Methods of cloning various dsRNA sequences into expression vectors, as well as expression vectors which can be adapted for use as described herein, are well known in the art, see, e.g. Schwab et al. 2006 Plant Cell 18: 1121-1133; Fraser. AfCS Reports 2004; Atayde et al. Mol Biochem Parasitol 2012 184:55-8: Kruhn et al. Cell Cycle 2009 8:3349-3354; and Timmons et al. Gene 2001 263: 103-112; which are incorporated by reference herein in their entireties.
[00107] In some embodiments, a vector for use in the methods and/or compositions described herein can comprise at least one constitutive promoter operably linked to at least one of the siRNA- binding polypeptide or the dsRNA multiple cloning site. In some embodiments, a vector for use in the methods and/or compositions described herein can comprise at least one inducible promoter operably linked to at least one of the siRNA-binding polypeptide or the dsRNA multiple cloning site.
[00108] Aspects of the technology described herein further relate to kits comprising the compositions described herein and kits for practicing the methods described herein.
[00109] In some embodiments, the technology described herein relates to a kit comprising a bacterial cell as described herein, e.g. a bacterial cell comprising at least a siRNA-binding polypeptide and a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially complementary to a target RNA as described herein.
[00110] In some embodiments, the technology described herein relates to a vector for use in the methods and compositions of the present technology, as described herein. In some embodiments, a kit for the production of one or more species of siRNA can comprise a vector comprising (a) a nucleic acid encoding a siRNA-binding polypeptide and (b) a dsRNA cloning site. In some embodiments, a kit for the production of one or more species of siRNA can comprise two vectors; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and wherein the second vector comprises a dsRNA cloning site. In some embodiments, the dsRNA cloning site can further comprise a dsRNA; wherein the dsRNA comprises a nucleic acid sequence substantially
complementary to a target RNA. In some embodiments, the kit can further comprise a bacterial cell.
[00111] In some embodiments, a kit for the production of one or more species of siRNA can comprise a bacterial cell comprising a siRNA-binding polypeptide and a vector comprising a dsRNA cloning site. In some embodiments, the bacterial cell can comprise a nucleic acid encoding a siRNA- binding polypeptide. In some embodiments, the nucleic acid encoding a siRNA-binding polypeptide can be a part of an expression vector, a plasmid, a naked nucleic acid, and/or the bacterial genome.
[00112] In some embodiments of a kit as described herein, the siRNA-binding polypeptide can comprise a purification tag. In some embodiments of a kit as described herein, the siRNA-binding polypeptide can be encoded by a nucleic acid. In some embodiments of a kit as described herein, the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA can be a portion of a vector. In some embodiments of a kit as described herein at least one of the siRNA-binding polypeptide or the dsRNA can be constitutively expressed. In some embodiments of a kit as described herein, at least one of the siRNA-binding polypeptide or the dsRNA can be inducibly expressed.
[00113] In some embodiments of a kit as described herein, the bacterial cell can express a siRNA- generating polypeptide. In some embodiments of a kit as described herein, the cell can be an Escherichia coli cell.
[00114] In some embodiments, the technology described herein relates to a kit comprising a library of siRNA species as described herein.
[00115] In some embodiments, the compositions and methods described herein can be used to test the efficacy of one or more siRNA species, and/or for the screening of a siRNA library. [00116] In some embodiments, the efficacy of one or more siRNA species can be assessed in cultured mammalian cells. Methods of targeting mammalian cells with inhibitory RNAs via bacterial invasion are known in the art, see, e.g. Zhao et al. Nature Methods 2005 2:967-973; which is incorporated by reference herein in its entirety. In some embodiments, a bacterial cell for use in such an assay can comprise a polypeptide or nucleic acid encoding a polypeptide which can bind to a mammalian cell surface receptor, (e.g. the invasin (inv) gene of Yersinia psuedotuberculosis which binds the integrin receptor of mammalian cells. In some embodiments, a bacterial cell for use in such an assay or screen can comprise a mutation reducing the ability of the cell to synthesize or maintain the cell wall, (e.g. deletion of the asd gene of E. coli, thereby rendering the cell a diaminopimelic acid (DAP) auxotroph). Reducing the ability of the cell to synthesize or maintain the cell wall can make the cell susceptible to lysis or degradation after it enters a mammalian cell, thereby releasing inhibitory RNAs (e.g. in the methods described herein, siRNAs) into the mammalian cell.
[00117] In some embodiments, the efficacy of one or more siRNA species can be assessed in vivo in C. elegans. dsRNAs readily cross cell membranes in C. elegans, and a number of protocols are known for conducting RNAi in C. elegans, including bacterial feeding assays (see, e.g. Timmons, L., and A. Fire. Nature 1998 395:854 and Lehner et al. Protocol Exchange 2006 159; which are incorporated by reference herein in their entireties.
[00118] In some embodiments, the efficacy of one or more siRNA species can be assessed by first isolating the one or more species of siRNA from the bacterial cells and then contacting a cell and/or organism with the one or more species of siRNA. Methods of introducing ribonucleic acids, and in particular, ribonucleic acids which cause RNAi into various cells and organisms are well known in the art (see, e.g. Sioud, M. "siRNA and miRNA Gene Silencing" Humana Press: 2011 ; "Gene Silencing by RNA Interference" Sohail, M. ed. CRC Press: 2004: each of which are incorporated by reference herein in their entireties). Examples of cells and/or organisms suitable for use in such methods include cultured cells (e.g. mammalian cells or human cells), primary cells, diseased cells (e.g.
cancerous cells), C. elegans, and Danio rerio.
[00119] The efficacy of one or more siRNA species can be assessed by screens, selections, and/or by assays. High throughput methods of screening siRNA libraries are known in the art, e.g.
phenotype screens, automated cell and worm processing, etc. The appropriate method of determining the efficacy of one or more siRNA species can be dependent upon the nature of the target RNA, e.g. siRNA species specific for target RNAs which control reproduction in C. elegans can be screened by examining the rate and success of reproduction of worms in the presence of the siRNAs.
[00120] In some embodiments, libraries of siRNA species as described herein, comprising siRNA species targeting a number of different target RNAs can be used in phenotypic screens to identify target RNAs associated with a particular phenotype (e.g. siRNAs which perturb a particular developmental process or which slow the progression of a disease). Phenotypic screens can comprise the assays described above for determining efficacy, e.g. mammalian cell invasion assays. In some embodiments, phenotypic screens can involve high-throughput assays.
[00121] In one aspect, described herein is a therapeutic agent comprising a siRNA species or population of siRNA species isolated from a bacterial cell as described herein and/or produced according to the methods described herein. According to the methods described herein, a dsRNA comprising a nucleic acid substantially complementary to a target RNA can be provided to a bacterial cell herein, and a siRNA species and/or population of siRNA species which can be used to reduce the expression of the corresponding target RNA can be produced. In some embodiments, target RNAs can be disease-associated RNAs, i.e. RNAs whose overexpression is associated with the cause, progression, or maintenance of a disease state, e.g. oncogenes. In some embodiments, target RNAs can be RNAs originating from a pathogenic organism, e.g. the target RNAs can comprise sequences of viral, bacterial, fungal, and/or parasitic origin. In some embodiments, target RNAs can be viral RNAs and/or RNAs produced from viral genomic material. In some embodiments, a siRNA species and/or population of siRNA species which can be used to reduce the expression of the target RNA can be produced according to the methods described herein and administered to a subject in need of a reduction of the level of expression of the target RNA. In some embodiments, a single siRNA species can be administered. In some embodiments, a population of siRNA species can be administered. As demonstrated in the Examples herein, a population of siRNA species can have increased efficacy and a lower likelihood of off-target effects as compared to a single siRNA species. In some embodiments, multiple populations of siRNA species can be administered, i.e. multiple target RNAs can be silenced. In some embodiments, the technology described herein relates to a pharmaceutical composition comprising a bacterial cell, siRNA species, and/or population of siRNA species according to the methods and compositions described herein. In some embodiments, the technology described herein relates to the use of a bacterial cell, siRNA species, or population of siRNA species according to the methods and compositions described herein in the manufacture of a medicament. Methods of preparing medicaments comprising RNA molecules, e.g. siRNAs, are known in the art, (see e.g. Oh and Park. Advanced Drug Delivery Reviews. 2009 61 :850-62; which is incorporated by reference herein in its entirety).
[00122] It is contemplated that the siRNA technology described herein, as well as the methods and compositions relating thereto, can be applied to gene silencing applications in any cell and/or organism comprising siRNA machinery. Non-limiting examples include gene silencing applications humans, non-human animals, livestock species, insects (e.g. honeybees), plants, crop plants, etc. In some embodiments, the gene silencing can be for therapeutic purposes. In some embodiments, the gene silencing can be for agricultural purposes, e.g. to treat agricultural diseases in animals and/or crops or to increase yields in animals and/or crops.
[00123] All patents and other publications; including literature references, issued patents, published patent applications, and co-pending patent applications; cited throughout this application are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the technology described herein. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.
[00124] The description of embodiments of the disclosure is not intended to be exhaustive or to limit the disclosure to the precise form disclosed. While specific embodiments of, and examples for, the disclosure are described herein for illustrative purposes, various equivalent modifications are possible within the scope of the disclosure, as those skilled in the relevant art will recognize. For example, while method steps or functions are presented in a given order, alternative embodiments may perform functions in a different order, or functions may be performed substantially concurrently. The teachings of the disclosure provided herein can be applied to other procedures or methods as appropriate. The various embodiments described herein can be combined to provide further embodiments. Aspects of the disclosure can be modified, if necessary, to employ the compositions, functions and concepts of the above references and application to provide yet further embodiments of the disclosure. Moreover, due to biological functional equivalency considerations, some changes can be made in protein structure without affecting the biological or chemical action in kind or amount. These and other changes can be made to the disclosure in light of the detailed description. All such modifications are intended to be included within the scope of the appended claims.
[00125] Specific elements of any of the foregoing embodiments can be combined or substituted for elements in other embodiments. Furthermore, while advantages associated with certain embodiments of the disclosure have been described in the context of these embodiments, other embodiments may also exhibit such advantages, and not all embodiments need necessarily exhibit such advantages to fall within the scope of the disclosure.
[00126] The technology described herein is further illustrated by the following examples which in no way should be construed as being further limiting.
[00127] Some embodiments of the technology described herein can be defined according to any of the following numbered paragraphs:
1. A bacterial cell comprising a siRNA-binding polypeptide and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
2. The bacterial cell of paragraph 1, wherein the siRNA-binding polypeptide comprises a
purification tag. The bacterial cell of any of paragraphs 1-2, wherein the siRNA-binding polypeptide is encoded by a nucleic acid.
The bacterial cell of any of paragraphs 1-3, wherein the siRNA-binding polypeptide is selected from the group consisting of:
pi 9 polypeptide; tombusvirus pi 9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
The bacterial cell of any of paragraphs 1-4, wherein the dsRNA is greater than 21 nucleotides in length.
The bacterial cell of any of paragraphs 1-5, wherein the dsRNA is a hairpin RNA.
The bacterial cell of any of paragraphs 1-6, wherein the bacterial cell expresses an RNase III polypeptide.
The bacterial cell of any of paragraphs 1-7, wherein the bacterial cell expresses an RNase III polypeptide encoded by an exogenous nucleic acid sequence.
The bacterial cell of any of paragraphs 1-8, wherein the bacterial cell is an Escherichia coli cell.
The bacterial cell of any of paragraphs 1-9, wherein at least one of the siRNA-binding polypeptide and the dsRNA are constitutively expressed.
The bacterial cell of any of paragraphs 1-10, wherein at least one of the siRNA-binding polypeptide and the dsRNA are inducibly expressed.
The bacterial cell of any of paragraphs 1-11, wherein the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA is part of a plasmid.
The bacterial cell of any of paragraphs 1-12, wherein the dsRNA comprises nucleic acid sequences substantially complementary to a multiplicity of target RNAs.
A method of producing one or more siRNA species which can inhibit the expression of a target RNA, the method comprising:
culturing a bacterial cell of any of paragraphs 1-13 under conditions suitable for the production of siRNAs.
The method of paragraph 14, further comprising a second step of isolating the siRNA-binding polypeptide and eluting the siRNAs bound to the siRNA-binding polypeptide.
The method of any of paragraphs 14-15, further comprising purifying the siRNAs eluted from the siRNA-binding polypeptide by HPLC.
The method of any of paragraphs 14-16, further comprising contacting the cell with one or more modified nucleotides before or during the culturing step.
A pharmaceutical composition comprising a siRNA produced according to the method of any of paragraphs 14-17.
The composition of paragraph 18, further comprising a population of siRNA species. A pharmaceutical composition comprising a siRNA isolated from a bacterial cell of any of paragraphs 1-13.
The composition of paragraph 20, further comprising a population of siRNA species.
The use of a siRNA produced according to the method of any of paragraphs 14-17 in the production of a medicament.
The use of a siRNA isolated from a bacterial cell of any of paragraphs 1-13 in the production of a medicament.
A vector comprising;
a nucleic acid encoding a siRNA-binding polypeptide; and
a dsRNA cloning site.
The vector of paragraph 24, wherein the dsRNA cloning site comprises at least one restriction enzyme site and can accept the insertion of at least one nucleic acid sequence such that a dsRNA is encoded and can be expressed.
A vector comprising:
a nucleic acid encoding a siRNA-binding polypeptide; and
a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
The vector of any of paragraphs 24-26, wherein the siRNA-binding polypeptide is selected from the group consisting of:
pi 9 polypeptide; tombusvirus pi 9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
The vector of any of paragraphs 24-27, wherein the vector is a plasmid.
The vector of paragraph 28, wherein the plasmid further comprises a bacterial origin of replication.
A library of siRNA species, the library comprising:
a plurality of clonal bacterial cell populations;
wherein each clonal population is comprises bacterial cells of any of paragraphs 1-13. A library of siRNA species, the library comprising:
a plurality of populations of siRNAs;
wherein each population of siRNAs is obtained according to the methods of any of paragraphs 14-17.
The library of paragraph 31, wherein each population of siRNAs binds to a single target RNA.
A kit comprising a bacterial cell of any of paragraphs 1-13.
A kit for the production of one or more species of siRNA, the kit comprising;
a bacterial cell comprising an siRNA-binding polypeptide; and at least one vector comprising a dsRNA cloning site.
A kit for the production of one or more species of siRNA, the kit comprising:
a bacterial cell comprising an siRNA-binding polypeptide; and
at least one vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
A kit comprising the vector of any of paragraphs 24-29.
A kit for the production of one or more species of siRNA, the kit comprising two vectors; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and
wherein the second vector comprises a dsRNA cloning site.
A kit for the production of one or more species of siRNA, the kit comprising two plasmids; wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and
wherein the second vector comprises a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
The kit of any of paragraphs 33-38, wherein at least one vector is a plasmid.
The kit of paragraph 39, wherein the plasmid further comprises a bacterial origin of replication.
The kit of any of paragraphs 33-40, wherein the kit further comprises a bacterial cell.
A kit for the production of one or more species of siRNA, the kit comprising;
a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA cloning site.
A kit for the production of one or more species of siRNA, the kit comprising;
a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
The kit of any of paragraphs 33-43, wherein the siRNA-binding polypeptide comprises a purification tag.
The kit of any of paragraphs 33-44, wherein the siRNA-binding polypeptide is encoded by a nucleic acid.
The kit of any of paragraphs 41-45, wherein the bacterial cell expresses an RNase III polypeptide.
The kit of any of paragraphs 41-46, wherein the cell is an Escherichia coli cell.
The kit of any of paragraphs 33-47, wherein at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to a constitutive promoter.
The kit of any of paragraphs 33-48, wherein at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to an inducible promoter. 50. The kit of any of paragraphs 33-49, wherein the DNA encoding at least one of the siRNA- binding polypeptide or the dsRNA is part of a plasmid.
51. A kit comprising the library of any of paragraphs 30-32.
EXAMPLES
[00128] RNA interference (RNAi) by double-stranded (ds) small interfering RNAs (siRNA) suppresses gene expression by inducing the degradation of mRNAs bearing complementary sequences1'2. Endogenous siRNAs (perfectly paired dsRNAs -21-25 nt in length) play an important role in host defense against RNA viruses and in transcriptional gene silencing in plants and may have similar functions in other eukaryotes3. Transfection of synthetic siRNAs into eukaryotic cells4'5 to silence genes has become an indispensable tool to investigate gene function, and siRNA-based therapy is being developed to knockdown genes implicated in disease6. Although bacteria expressing sense and antisense sequences can be fed to worms to knock down individual genes7, no one has used living organisms to produce highly active, purified siRNAs. Described herein is a method to produce highly potent siRNAs from E. coli ectopically expressing pi 9, a siRNA binding protein, which stabilizes siRNA-like species generated by bacterial RNase III.
[00129] The most common method to make siRNA is chemical synthesis4'5. Effective siRNA sequences are predicted using computer algorithms. siRNAs can also be made from transcribed longer dsRNAs by in vitro biochemical processing by RNase III family enzymes8'9. In the latter case, the resulting siRNAs contain many sequences against one target, which sometimes can be more effective than any one sequence10, and pools of siRNAs often have fewer off-target effects on genes bearing partially complementary sequences11. While gene knockdown by transfection of siRNAs is usually transient, short hairpin RNA construct, delivered by plasmid or lentivirus, is commonly used to achieve stable gene silencing.
[00130] pl9, an RNAi suppressor protein encoded by the plant RNA virus tombusvirus12, selectively binds to -21 nt siRNAs, including those targeting the virus13. The pi 9 dimer binds to the -19 nt duplex region of an siRNA in a sequence-independent manner14'15. It was originally planned to enrich for endogenous siRNAs in mammalian cells using pi 9 coupled to magnetic beads15. As a negative control, pl9 beads were incubated with total RNA isolated from E. coli, an organism that supposedly lacks the RNAi machinery, that was transformed or not with a pcDNA3.1+ plasmid in which pl9 was cloned after the CMV immediate-early promoter. Surprisingly pi 9 beads pulled down -21 nt dsRNAs from RNA of both human T-cells (ACH2 cell line) and the transformed E. coli cells (Figure 1 A). Although the CMV promoter is mostly used for efficient expression of genes in mammalian cells, E. coli harboring pcDNA3.1+ plasmids encoding FLAG-tagged TREXl or pi 9 gene expressed their respective proteins (Figure IB). When total RNA isolated from E. coli transformed with empty vector or vectors encoding pi 9 or TREXl was separated on SYBR Gold-stained denaturing polyacrylamide gels, a distinctive -21 nt band was evident only in pl9-expressing E. coli (Figure IB). These data indicate that pi 9 protein expression may have stabilized a cryptic siRNA-like RNA species in E. coli. In Listeria monocytogenes, a Gram-positive bacterium, expression of pi 9 also allowed the detection of -21 nt small RNAs (Figures 5A-5B).
[00131] To determine if the small RNAs detected in E. coli depended on functional pl9, RNA was isolated from E. coli expressing WT pl9, or pl9 mutants that disrupted siRNA binding14'16 (Figure 1C). The -21 nt dsRNA band was more prominent in bacteria expressing WT pl9. Thus siRNA- binding to pi 9 promotes the accumulation of siRNA-like RNAs in E. coli. Next the nuclease responsible for making small RNAs was sought. The most likely candidate was RNase III, an ancestor of eukaryotic Dicer, responsible for the final step of siRNA biogenesis17. E. coli RNase III is known to generate siRNA-sized dsRNAs from longer dsRNAs in vitro9. pl9-expressing plasmids were used to transform two RNase III mutant strains, rncl4li and rnc3819 (Figure ID). In both mutant strains, pi 9 beads failed to pull down any visible small RNAs. Furthermore restoration of RNase III expression in HT115(DE3), a rncl4 strain, also restored the pl9-dependent small RNAs (Figure IE), providing support for the hypothesis that RNase III is responsible for generating these small RNAs in E. coli. Thus, accumulation of these bacterial small RNAs depends on ectopic pi 9 and bacterial RNase III.
[00132] It was next asked whether small RNAs generated in pl9-expressing E. coli behave like siRNAs and can be used for gene knockdown in mammalian cells, pi 9 was cloned into the pGEX-4T- 1 plasmid to express a GST-pl9 fusion protein with a C-terminal His tag (Figure 2A). A T7 promoter driving expression of a hairpin RNA that contains a target sequence was inserted immediately after. To develop the method, a hairpin was designed that encoded full-length EGFP {EGFPFL). The expression of the GST-pl9-His fusion protein and hairpin RNA were both induced by IPTG. The GST-pl9-His protein was captured by Nickel (Ni) affinity chromatography and 0.5% SDS was used to selectively elute pl9-bound RNAs that were predominantly -21 nt long (Figures 2B and 6A-6D). Small RNAs were further purified from other longer RNAs by anion exchange HPLC. To verify that these bacterial small RNAs are double-stranded, they were treated with a variety of nucleases. Like chemically synthesized siRNAs, bacterial small RNAs were sensitive to RNase A, but were insensitive to enzymes that digest ssRNA or DNA (Xrnl, RNase Tl, exonuclease T (Exo T), exonuclease I (Exo I), or DNase Turbo (Figure 2C). Next bacterial small RNAs, purified from E. coli expressing pi 9 and the EGFPFL hairpin and transfected into HeLa cells stably expressing dlEGFP (HeLa-dlEGFP), were loaded into the RNA-induced silencing complex (RISC) by
immunoprecipitation with a pan-Argonaute (Ago) antibody (Figure 2D). RNAs that precipitated with anti-Ago were -21 nt long and hybridized to an EGFP probe, but no small RNA precipitated with control mouse IgG. Thus bacterial small RNAs were similar to synthetic siRNA in chemical composition and were incorporated into the RISC. These small RNAs were named 'pro-siRNAs' for prokaryotic siRNAs. [00133] Since pro-siRNAs had properties of siRNAs, whether pl9-captured EGFP pro-siRNAs induce gene knockdown was tested. qRT-PCR and flow cytometry were used to compare mRNA and protein knockdown, respectively, of dlEGFP in HeLa-dlEGFP cells transfected with a synthetic EGFP siRNA or pro-siRNAs purified from E. coli expressing pi 9 and hairpins of either full length EGFP (EGFPFL) or a 100 nt fragment that overlapped with the EGFP siRNA sequence (EGFP 100). Both EGFPFL and EGFP 100 pro-siRNAs knocked down EGFP expression more effectively than equimolar concentrations of siRNA (Figures 2E and 7A). pro-siRNAs made from the plasmid without or with only half of the EGFP hairpin could not silence EGFP effectively (Figure 7B). As expected, silencing by pro-siRNA was Dicer-independent because EGFPFL pro-siRNA still functioned in Dicer-deficient HCT116 cells20 and recombinant Dicer protein did not further process pro-siRNAs in vitro (Figures 8A-8B).
[00134] To test the effectiveness of pro-siRNA knockdown of endogenous and viral genes, we used convenient restriction sites to clone and express hairpins from the coding regions of LMNA (which encodes two splice variant products, lamin A and lamin C), PLKl, TP 53 and HIV vif (viral infectivity factor) and gag (capsid antigen) to purify pro-siRNAs. The resulting hairpins contained 200-579 nt of each sense and antisense sequence (523 nt for LMNA, 299 nt for PLKl, 300 nt for TP53, 579 nt for vif200 and 500 nt for gag). The HPLC-purified pro-siRNAs for each gene contained a few different sized species that migrated close to the 21 nt marker on both native and denaturing polyacrylamide gels (Figure 2F). For LMNA and PLKl pro-siRNAs, a minor RNA band migrated at -25 nt. Next the extent of knockdown of endogenous genes (LMNA, TP 53, PLKl) by pro-siRNAs and commercially available siRNAs (LMNA and TP53 siRNAs were from a single sequence; PLKl siRNAs were a pool of 4 siRNAs and were chemically modified by proprietary methods for enhanced stability and reduced off-target effects21) in HeLa-dlEGFP and HCT116 cells were compared. The extent of gene knockdown was similar between siRNA and pro-siRNA transfected at 4 nM (Figure 3A). Since knocking down PLKl causes death of dividing cells22, viable cells were counted for 3 d following transfection with PLKl or control siRNAs and pro-siRNAs (Figure 3B). To more closely evaluate the potency of pro-siRNAs, dose response experiments comparing transfection of pro- siRNAs (0.2, 2, 20 nM) targeting LMNA, TP53 and PLKl with five commercial siRNAs for each gene (four siRNAs from Dharmacon, of which the PLKl siRNAs were chemically modified for enhanced RISC uptake or stability by proprietary methods, and one siRNA sequence chosen based on published effectiveness) were performed (Figure 9). The potency of the commercial siRNAs varied, as best evaluated at the lowest concentration. The pro-siRNAs, whose sequences were not optimized, achieved similar gene knockdown as the commercially optimized siRNAs. At a concentration of 2 nM, each pro-siRNA achieved knockdown of -90%. Because siRNA design algorithms are imperfect, identifying a potent siRNA usually requires test of several sequences which could be costly and time consuming. pro-siRNAs might circumvent the need to test multiple sequences to identify a single potent siRNA. [00135] To examine potential toxicity of pro-siRNAs, growth was compared over 3 d in HeLa- dlGFP and HCT116 cells after transfection with either a negative control siRNA or EGFP pro-siRNA (Figure 3B). Their growth curves were not significantly different. To compare the effectiveness of gene knockdown by pro-siRNAs and siRNAs, cell proliferation was examined after knocking down PLK1, which kills dividing cells23. PLK1 siRNAs and pro-siRNAs both dramatically reduced viability with indistinguishable kinetics.
[00136] As another test of pro-siRNA function, the effect of knocking down the HIV accessory gene vif on in vitro propagation of HIV infection23 was examined, vif, which targets the host restriction factor APOBEC3G for ubiquitylation and degradation, is not needed for the initial round of HIV replication, but is required to spread the infection to new cells by preventing APOBEC3G packaging into budding virions. The efficacy of the pro-siRNAs was compared with two validated siRNAs23'24. As expected, siRNAs and pro-siRNAs targeting vif 'did not alter the percentage of initially infected HeLa-CD4 cells (data not shown), but did suppress vif gene expression and inhibit subsequent rounds of infection, assessed in the TZM-bl luciferase reporter cell line (Figure 3D). Transfection of vif pro-siRNAs resulted in much lower levels of vz/mRNA in HeLa-CD4 cells and HIV tat-driven luciferase activity, compared to transfection with either or both vz/siRNAs. Thus vif pro-siRNAs were superior to previously used siRNAs in inhibiting HIV spread in vitro.
[00137] One major obstacle to using RNAi to suppress HIV or other viruses is sequence diversity. Because pro-siRNAs target many sequences within a gene, pro-siRNAs directed against a viral gene can have broader activity against diverse viral strains than siRNAs and can also be less likely to generate siRNA-resistant mutants. Previous attempts to identify an siRNA against HIV-1 clade B gag that could inhibit viral isolates from other clades were unsuccessful24. A sequence that protected against infection with all clade B viruses was tested, but no single sequences which were well enough conserved were found that also protected against other clades. To investigate whether pro-siRNAs might have broader activity than the best clade B sequence, gag pro-siRNAs were engineered using hairpins with 200 and 500 nt long stems from the gag coding region of clade B HIV-IIIB virus. The gagB200 and gagB500 pro-siRNAs more potently suppressed HIV-IIIB, than the previous gag siRNA (Figure 2D). More importantly, unlike the gag siRNA, both gag pro-siRNAs knocked down gag mRNA and inhibited viral spread in vitro for UG29 (clade A) and ΓΝ22 (clade C) viruses, although they worked slightly less effectively than against IIIB virus. These data indicate that pro-siRNAs could be particularly beneficial for targeting heterogeneous and rapidly evolving viral genes.
[00138] Because mammalian cells are sensitive to bacterial endotoxin, which stimulates off-target innate immunity via Toll-like receptor signaling, whether purified pro-siRNAs are contaminated with endotoxin was assessed. Although SDS-eluted pro-siRNAs contained significant amounts of endotoxin, assayed by Limulus amoebocyte lysate (LAL) assay, HPLC purified pro-siRNAs, even at concentrations as high as 320 nM, were below the limit of detection (0.25 EU/ml) (Table 1).
Endotoxin contamination was tested for by assaying for induction of mRNA expression of the proinflammatory cytokines TNFA, IL6, IL8 and IL12, measured 4 hr later by qRT-PCR in highly endotoxin-sensitive monocyte-derived human macrophages (Figure 10A). Incubation with HPLC- purified vz/pro-siRNAs (320 nM) did not trigger cytokine gene expression. Thus purified pro-siRNAs did not contain significant amounts of immunostimulatory endotoxin. Next MDMs were transfected with a few siRNAs and pro-siRNAs (at 20 nM) to test immune response mediated by endogenous immune sensors (Figure 10B). siRNA and pro-siRNA against LMNA efficiently down regulated LMNA rriRNA, indicating these siRNAs were successfully transfected into MDMs. Comparing to siRNAs, pro-siRNAs did not trigger excessive activation of immune genes.
[00139] To ascertain the sequence composition of pro-siRNAs, pro-siRNAs were cloned and deep sequenced using a cloning method established for eukaryotic siRNAs (sequencing reads and alignment summary in Table 2). Most reads were concentrated between 20 and 22 nt (Figures 4A and 11). The majority of reads (on average -75%) aligned to the target sequence, plasmid backbone or the E. coli genome. The vast majority of aligned sequences (82-99%) originated from the target sequence (Figure 4B); consistent with the efficient gene knockdown they induced. Reads were generated from the entire target sequence, but were also concentrated at specific sites ('hot spots') (Figures 4C, 11, and 12A-E). There was some sequence strand bias for most of the hot spots (Figure 12A). Because the data (Figures 2C and 2F) strongly suggested that pro-siRNAs are double stranded, it was possible that strand bias may have been due to differences in ligation efficiency during cloning, a well-known problem25, rather than the presence of many single-stranded RNAs. To evaluate this further, forward and reverse DNA oligonucleotide probes (26-27 nt) were designed for three EGFPFL pro-siRNA hot spots and performed solution hybridization and native gel electrophoresis (Table 3 and Figure 12B). The relative intensity of hybridized bands was approximately equal for sense and antisense probes for each hot spot and were generally correlated with the number of reads from each hot spot (Figures 12C-12E). Thus, pro-siRNAs are mostly dsRNAs and the strand bias in the deep sequencing data likely reflects ligation bias during cloning.
[00140] To further investigate the hot spot pattern, siRNA profiles of two independent preparations of EGFPFL pro-siRNAs cloned using different sets of adapters were compared. The potency, size profile and sequence content of two EGFPFL pro-siRNAs were similar, but not identical. The most abundant hot spots were consistent in the 2 samples, but the strand bias changed with the adapters, consistent with cloning bias (Figure 13A-D). Without wishing to be bound by theory, hot spots could be due to intrinsic sequence preferences for RNase III cleavage or differences in stability or pl9 binding after cleavage. To determine whether 'hot spots' are determined by sequence differences at or close to the hot spot, hairpins of equal sizes were constructed from the 5' and 3' ends of the full length EGFP sequence. The pro-siRNAs generated from the two halves yielded mostly identical hot spots to the corresponding hot spots in EGFPFL pro-siRNAs (Figure 13E). Thus hot spots seem to be determined by local sequence differences. However a basic bioinformatic analysis of sequence motif or preferred base for the hot spots was inconclusive (data not shown). E. coli RNase III might process dsRNA into siRNA-sized small RNAs in vivo through a mechanism that differs from Dicer27, whose cleavage of a long dsRNA results in phased and evenly distributed sequences along a target gene.
[00141] Because pro-siRNAs contained non-targeting sequences derived from the plasmid or E. coli genome, possible off-targeting effects26 were investigated. To evaluate off-targeting, RNA expression profiles were compared by RNA deep sequencing of HeLa-dlEGFP cells transfected with 4 nM of negative control or EGFP siRNA or EGFPFL or EGFPIOO pro-siRNAs (sequencing reads and alignment summary in Table 2). Tophat and Cufflinks were used to analyze the data and plotted volcano plots of all annotated transcripts (fold change versus p value, Figure 4D). Comparing to EGFP siRNA, EGFPIOO pro-siRNA had higher number of significantly changed genes while EGFPFL pro-siRNA had less (Figures 4F and 14A). EGFPFL pro-siRNA also produced the least changes in long non-coding RNAs, a group of newly discovered gene regulators (Figures 14B-14C). EGFPIOO pro-siRNAs, made from a shorter hairpin (100 bp), contained higher proportion of plasmid and genomic sequences compared to other pro-siRNAs made from longer hairpins (200 to 720 bp, Figure 4B), which is likely the cause of higher off-target effect. These data indicate a plasmid containing longer sequences of the target gene could have fewer off-target effects. Gene expression profiles of cells transfected with LMNA siRNAs and pro-siRNAs were also compared by microarray. Consistent with the EGFP data, LMNA pro-siRNAs, made from a longer hairpin (523 bp), produced fewer number of significantly changed genes comparing to LMNA siRNA (Figures 4E, 4F, and 14D). The RNA profiling data also showed the target gene was always the most down regulated gene and pro-siRNAs consistently produced better knockdown than siRNA. Thus pro-siRNAs could be engineered to offer better knockdown and lower off-target effects compared to synthetic siRNAs. The significantly changed genes in each of these experiments were not enriched for innate immune genes30, confirming that the pro-siRNAs did not stimulate an innate immune response. Thus pro- siRNAs offer highly specific knockdown that is at least as good as synthetic siRNAs without the need to test multiple sequences.
[00142] It is demonstrated herein that bacteria can be genetically engineered to produce siRNAs that are highly effective and not toxic to mammalian cells. Specifically, it demonstrated herein is efficient knockdown of one exogenous gene {EGFP), two viral genes (vif and gag) and 3 host genes (PLKl, TP53, LMNA). Without wishing to be bound by theory, because pro-siRNAs are natural products of RNase III, they likely have favorable ends (e. g., 5'-phosphate, 3'-hydroxyl and 3' overhangs) for efficient loading by Ago into the RISC and do not activate cytosolic innate immune RNA sensors. An alternative strategy of producing pro-siRNAs that uses two plasmids - one to express pi 9 and the other to transcribe both sense and antisense strands of a target sequence - facilitates cloning and can also be used to produce efficient gene silencing (Figures 15A-15C).
[00143] Without much optimization an average yield of ~4 nmol (-42 μg) pro-siRNA per liter of E. coli culture was achieved. It is contemplated that the engineered plasmid or E. coli genome could potentially be further optimized to maximize yield and improve effectiveness. By way of non-limiting example, the yield of EGFPFL pro-siRNA could be doubled by overexpressing E. coli RNase III (Figure 16).
[00144] Generating pro-siRNAs for research purposes might be more cost effective than purchasing and testing multiple individual chemically synthesized siRNAs. pro-siRNAs, containing multiple sequences, might offer fewer off-target effects than individual siRNAs and could be harder for the target gene to escape silencing by mutation. On the other hand, chemical synthesis provides the opportunity for chemical modifications to increase potency, enhance stability and reduce off-target effects or couple fluorophores or targeting moieties. Such modifications might also be possible for pro-siRNAs, either by adding modified ribonucleotides to bacterial cultures during IPTG induction or by performing the same coupling reactions with purified pro-siRNAs as are used to modify siRNAs, respectively.
[00145] RNase Ill-deficient E. coli expressing dsRNAs can be fed to C. elegansli and bacteria- derived dsRNAs can be applied to plants to induce specific gene knockdown28. However, gene silencing requires host Dicer and, unlike for mammalian cells, is enhanced in these organisms by RNA-dependent RNA polymerases that can amplify small amounts of RNA. More recently, genetically engineered E. coli, designed to express an invasin to induce bacterial uptake and listeriolysin, to allow bacterial RNAs to escape from phagolysosomes, delivered dsRNAs into the cytoplasm of human cells through "trans-kingdom RNAi" technology29.
[00146] pro-siRNAs, described here, could become a valuable cost effective addition to existing RNAi techniques for both research and therapeutics. The method described herein for producing pro- siRNAs can easily be adopted and scaled-up in an industrial setting. It is contemplated that mammalian cDNA libraries could be used to generate pro-siRNA libraries, e.g. for siRNA screening pro-siRNAs, generated from longer hairpins containing multiple sequences, might offer fewer off- target effects than individual siRNAs and in the cases of virus infection or cancer might be harder for the target gene to escape from by mutation. On the other hand, chemical synthesis provides the opportunity for chemical modifications to increase potency, enhance stability and reduce off-target effects or to couple fluorophores or targeting moieties. Such modifications can be applied to pro- siRNAs, e.g. either by adding modified ribonucleotides to bacterial cultures during IPTG induction or by performing the same coupling reactions with purified pro-siRNAs as are used to modify siRNAs, respectively.
Methods
[00147] Bacterial strains and culture conditions. All E. coli strains used in this study are listed in Table 4. E. coli strain DH5a was used for cloning and for initial characterization of the siRNA-like RNA species. For recombinant protein expression and pro-siRNA production, T7 Express Iq (NEB), a BL21 -derived E. coli strain was used. Two mutants of RNase III, rnc- 74::DTnlO (TetR) and Ornc-38 (KanR) were utilized. These were moved by PI transduction from parent strains HT115(DE3) 18 and SK7622 into E. coli strain MG1655 AlacZYA (also referred as MG1655 Mac). All E. coli strains were cultured in LB broth, Lennox (BD) at 37°C with shaking at 250 rpm and antibiotics when required were used at the following concentrations; carbenicillin (100 μg/ml), kanamycin (50 μg/ml), spectinomycin (50 μg/ml), tetracycline (12.5 μg/ml).
[00148] Listeria monocytogenes strain 10403S was cultured in brain-heart infusion medium (BD Biosciences) at 30°C. Transformation of bacterial cells was performed as previously described32.
[00149] Genes and plasmids. The pi 9 gene used in this study was cloned from Tomato bushy stunt virus. All plasmids are listed in Table 5. To produce pi 9 in E. coli, pcDNA3.1+ (Invitrogen) was used to express the pi 9 protein with a C-terminal FLAG tag (pcDNA3.1-pl9-FLAG) or an N-terminal His tag (pcDNA3.1-His-pl 9). Plasmid pcDNA3.1 -TREX 1 -FLAG encodes a C-terminal FLAG-tagged TREX1 protein. To express pi 9 in L. monocytogenes, pLIV-l-His-pl9 plasmid was used, which encodes pi 9 with an N-terminal His tag cloned in pLIV-1 plasmid (gift of Darren Higgins, Harvard Medical School). E. coli RNase III with an N-terminal FLAG was cloned in pcDNA3.1+ and pCDF- lb (Novagen) plasmids.
[00150] Two strategies were used for pro-siRNA production in E. coli. In one approach pl9-His was fused to GST in pGEX-4T-l (to express GST-pl9-His fusion protein). On the same plasmid we cloned a hairpin RNA expressing cassette consisting of inverted repeat separated by a 32 bp linker downstream of a T7 promoter. A scheme of the resulting plasmid, pGEX-4T-l -pl9-T7, is showed in Figure 13A-13D. The hairpin RNA sequences were: EGFPFL, the entire 720-bp EGFP coding sequence (from pEGFP-Nl, Clontech); EGFP100, a 100 bp from nt 219 to 318; EGFP Hotspot-1 360 bp from nt 1 to 360; EGFP Hotspot-2 360 bp from nt 361 to 720; LMNA (NM 005572.3), 523 bp from nt 267 to 789; TP53 (NM_000546.5), 301 bp from nt 376 to 676; PLK1 (NM_005030.3), 299 bp from nt 92 to 390; vz/(K03455), the entire 579-bp; gag (K03455), gagB200: 200 bp from nt 1183 to 1382, gagB500 500 bp from nt 1004 to 1503. (Genbank entries listed; numbers refer to position with respect to the translation start site).
[00151] In another approach two compatible plasmids were used for pro-siRNA production. The GST-pl9-His protein was cloned under the control of the T7 promoter in pRSF-lb (Novagen) or pCDF-lb to generate pRSF-GST-pl9-His and pCDF-GST-pl9-His. The second plasmid is a L4440 plasmid encoding the entire EGFP coding sequence (L4440-EGFP).
[00152] All cloning was performed using PCR and standard techniques. All primers (with information for restriction enzyme sites) are listed in Table 6.
[00153] Cells. HeLa-dlEGFP, HCT116, HCT116 Dicer '-, HeLa-CD4 TZM-bl,
U87.CD4.CXCR4 and U87.CD4.CCR5 cells were cultured in DMEM medium (Invitrogen) supplemented with 10% heat-inactivated fetal bovine serum (FBS). ACH2 cells (human leukemia T cell line CEM latently infected with HIV-1) were cultured in RPMI medium (Invitrogen)
supplemented with 10% heat-inactivated fetal bovine serum (FBS). For assays using primary monocyte-derived human macrophages (MDM), monocytes were isolated from blood of a healthy donor by Ficoll-Paque Plus (GE Healthcare) density separation. Monocytes were plated on
PRIMARIA plates (FALCON) in RPMI medium (Invitrogen) supplemented with 10% heat- inactivated human serum and adherent cells were cultured for 5 d to allow differentiation into macrophages.
[00154] RNA isolation and qRT-PCR. Total RNA was isolated from 3 ml of E. coli stationary phase culture with 1 ml Trizol reagent (Invitrogen) following the manufacturer's protocol. RNA from human cells was collected in Trizol and extracted according to the manufacturer's protocol. Total RNA (1 μg) was converted to cDNA using Superscript III Reverse Transcriptase (Invitrogen). For qRT-PCR, 10 μΐ reaction, containing SsoFast EvaGreen mastermix (Bio-Rad), appropriate primers (Table 4), and template cDNAs made from 10 ng RNA, was amplified on a Bio-Rad CFX 96 Thermal Cycler. All qRT-PCR data were normalized to the human GAPDH gene. qRT-PCR primers for human genes (Table 6) were selected from PrimerBank (available on the world wide web at
pga.mgh.harvard.edu/primerbank/).
[00155] siRNA isolation from total RNA using pl9 magnetic beads. pl9 magnetic beads were prepared at NEB as previously described15. To pull down siRNAs, 50 μg of total RNA (isolated from human or E. coli cells) was used following the manufacturer's protocol15.
[00156] His-tag purification of GST-pl9-His and bound pro-siRNA. GST-pl9-His was purified as follows. A fresh single transformant of T7 Express Iq containing pGEX-4T-l -pl9-T7 was used to inoculate 300 ml LB medium in a 1.5 L flask. When the OD60o reached 0.3-0.6, protein and pro- siRNA expression were induced by adding 0.5 mM IPTG for 1 hr. Cells were centrifuged and lysed in 10 ml lysis buffer (50 mM Phosphate buffer pH 7.0, 300 mM NaCl, 10 mM imidazole, 1% Triton X- 100, 1 mg/ml lysozyme) at 4°C for -30 min followed by sonication (Misonix S-4000) until the lysate was non-viscous. Following centrifugation the lysate was incubated with rotation with 1 ml Ni-NTA resin (Thermo Scientific) overnight at 4°C. The resin was washed with lysis buffer 4 times, each time for 10 min at 4°C with rotation. Bound GST-pl9-His was eluted in lysis buffer containing 300 mM imidazole at room temperature.
[00157] To purify pl9-bound pro-siRNA the procedure was as above until the final elution step when 500 μΐ 0.5% SDS was added for 10 min at room temperature with rotation. This step was repeated and both SDS eluates were combined and passed through a 0.22 μηι centrifuge filter (Corning) before HPLC purification on a Bio WAX NP5 anion exchange column (Agilent
Technologies). The HPLC buffers were: Buffer A, 25 mM Tris-HCl, 2 mM EDTA; Buffer B, 25mM Tris-HCl, 2 mM EDTA, 5 M NaCl. HPLC was initiated with a flow rate of 1 ml/min at 25°C. Elution was performed using a linear gradient of 0-10% Buffer B over 4 min, followed by 10% Buffer B for 6 min, and a second linear gradient of 10-25%) Buffer B over 15 min at a reduced flow rate of 0.5 ml/min. pro-siRNA eluted in the second gradient was collected by isopropanol precipitation.
[00158] Polyacrylamide gel electrophoresis (PAGE) of RNA. For denaturing electrophoresis of RNA, mini-sized pre-cast 15% polyacrylamide TBE-Urea gels (Invitrogen) were used. RNA samples were heated to 95°C for 5 min in Gel Loading Buffer II (Ambion) and then immediately placed on ice until gel loading. Electrophoresis was performed in a 70°C water bath (to ensure complete denaturation of siRNA) and gels were stained with SYBR Gold (Invitrogen). For analysis of E. coli total RNA, 20 μg samples of Trizol-isolated RNA were loaded. RNA size standards (miRNA marker, siRNA marker and Low Range ssRNA Ladder) were from NEB.
[00159] For native electrophoresis of RNA, mini-sized homemade 15% polyacrylamide TBE gels were used with the Bio-Rad Mini-PROTEAN Tetra Cell. RNA samples were prepared in Gel Loading Buffer II (Ambion) without heat denaturation and electrophoresis was performed at room temperature.
[00160] Nuclease sensitivity assay. The nucleases tested were: RNase A, RNase Tl, and Turbo DNase (all from Ambion), Xrnl, exonuclease T, and exonuclease I (all from NEB). For each assay, 200 ng of an unmodified synthetic negative control siRNA (GenePharma) and vz/pro-siRNA were used and assays were incubated in a 20 μΐ reaction volume using standard amounts of enzymes at 37°C for 1 hr. Treated RNAs were purified by phenol/chloroform extraction followed by isopropanol precipitation.
[00161] Test for endotoxin activity and immune activation in primary human monocyte-derived macrophages (MDM)/ RNA samples diluted in ddH20 to the indicated concentration were analyzed by the single vial Gel Clot LAL assay (detection limit 0.25 EU/ml, Lonza) following the
manufacturer's protocol. Lipopolysaccharide (LPS) from E. coli Ol l l :B4 (Sigma-Aldrich) was used as a positive control.
[00162] To test for cytokine gene activation, MDM plated in 24 well plates (lxlO5 cells/well) were incubated with medium containing RNA or LPS at the indicated concentration for 4 hr before harvesting RNA. siRNAs and pro-siRNAs were also transfected to MDMs at 20 nM using
Lipofectamine 2000 (Invitrogen) and total RNA were harvest at 24hrs after transfection.
[00163] 5' 32P labeling of RNA. RNA samples were dephosphorylated by Antarctic Phosphatase (NEB) for 30 min at 37°C in the presence of Murine RNase Inhibitor (NEB). The Antarctic
Phosphatase was deactivated by incubation at 65°C for 5 min and the RNA was end-labeled with γ-32Ρ ATP (PerkinElmer) and T4 Polynucleotide Kinase (NEB). Gels were exposed using a phosphorimager screen and visualized using a FLA-9000 Image Scanner (Fujifilm).
[00164] Small RNA northern blot. Northern blot for small RNAs was performed as previously described33. The EGFP specific sense probe was a 32P-UTP -internally labeled RNA prepared by in vitro transcription using T7 RNA polymerase (NEB) and a PCR-generated DNA template of the full- length EGFP gene that incorporated a T7 promoter.
[00165] siRNA transfection for testing RNA silencing efficiency. All siRNA transfections were performed using Lipofectamine 2000 following the manufacturer's protocol. Briefly, cells were plated in 24 well plates ( 1 x 105 per well) and the transfection complex (containing 1.0 ml Lipofectamine 2000 and siRNAs) was added directly to the medium. RNA and protein samples were isolated from cells 24 hr post-transfection. For the PLK1 cell killing experiment, cells were counted using a TC-10 automatic cell counter (Bio-Rad). The following siRNAs were used: ON-TARGETplus Non-targeting siRNA #4 (D-001810-04-05, Dharmacon), siGENOME Lamin A/C Control siRNA (D-001050-01-20, Dharmacon), Set of 4: siGENOME LMNA siRNA (MQ-004978-01 -0002, Dharmacon), ON- TARGETplus SMARTpool - Human PLK1 (L-003290-00-0005, Dharmacon), Set of 4 Upgrade: ON- TARGETplus PLK1 siRNA(LU-003290-00-0002, Dharmacon), Set of 4: siGENOME TP53 siRNA (MQ-003329-03-0002, Dharmacon), Negative control siRNA (NC siRNA, B01001, GenePharma), Positive control siRNA TP53 (B03001, GenePharma), custom EGFP siRNA (sense,
GGCUACGUCCAGGAGCGCACC (SEQ ID NO: 114); antisense,
UGCGCUCCUGGACGUAGCCUU (SEQ ID NO: 115)), custom vz/siRNA-123 (sense,
GUUCAGAAGUACACAUCCCT (SEQ ID NO: 116); antisense,
GGGAUGUGUACUUCUGAACTT (SEQ ID NO: 117)) and custom siRNA-224 (sense,
CAGAUGGCAGGUGAUGAUUGT (SEQ ID NO: 118); antisense,
AAUCAGCACCUGCCAUCUGTT (SEQ ID NO: 119)), custom gag siRNA: (sense,
GAUUGUACUGAGAGACAGGCU (SEQ ID NO: 120); antisense,
CCUGUCUCUCAGUACAAUCUU (SEQ ID NO: 121)).
[00166] RISC Immunoprecipitation. Cells (3x106) were transfected with 4 nM NC siRNA or EGFPFL pro-siRNAs. After 24 hours cells were scraped from the plate in 2 ml lysis buffer (150 mM KC1, 25 mM Tris-HCl pH 7.5, 2 mM EDTA, 0.5 mM DTT, 1% NP-40 and Roche Complete Protease Inhibitor Cocktail). Cells were then mechanically disrupted for 1 min using a micro-MiniBeadbeater (BioSpec). The cell lysate was incubated at 4°C with rotation for 1 hr to ensure complete lysis. IP was performed by adding anti-Ago (2A8) antibody (Millipore, MABE56) or mouse total IgG (Jackson Labs) at 1 : 100 dilution together with 30 μΐ protein G Dynabeads (Invitrogen) and samples were rotated at 4°C overnight. After washing 4 times in lysis buffer, precipitated RNAs were isolated using Trizol reagent from 90% of the reaction mix, while 10% was saved for immunoblot input.
[00167] Western Immunoblot. Protein samples were prepared by heating cells to 95°C for 5 min in IX SDS loading buffer before SDS-PAGE. Immunoblot was performed using SNAP i.d. Protein Detection System (Millipore) following the manufacturer's protocol. Antibodies and their dilutions were: anti-FLAG (M2) 1 : 1,000 (Sigma- Aldrich, F1804), anti-His tag 1 :500 (Covance, MMS-156P), anti-PLKl 1 : 100 (Santa Cruz, sc-17783), anti-LaminA/C 1 : 1,000 (Santa Cruz, sc-7292), anti-p53 (DO-1) 1 :500, (Santa Cruz, sc-126), anti-beta-Tubulin 1 : 10,000 (Sigma- Aldrich, T5168), anti-Ago (2A8) 1 : 1,000 (Millipore, MABE56). Horseradish peroxidase conjugated anti-mouse or anti-rabbit IgG secondary antibodies were used at 1 :5,000 dilution followed by incubating the membranes in SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific).
[00168] Solution hybridization and native gel electrophoresis assay. DNA oligonucleotides purchased from IDT were PAGE purified. Purified DNA oligonucleotides (10 pmol) were end-labeled with γ-32Ρ ATP by T4 Polynucleotide Kinase (NEB) and 2 pmol was then mixed with 5 ng of pro- siRNAs in buffer containing 20 mM Tris-HCl pH 7.9, 100 mM NaCl and 2 mM EDTA. Samples were heated to 80°C for 10 min and allowed to cool to room temperature. A fraction of the sample was separated on a native 15% polyacrylamide gel. The gel was directly exposed to a phosphorimager screen. Multi-gauge software (Fujifilm) was used for image quantification.
[00169] siRNA library preparation, deep sequencing, and data analysis. siRNAs were cloned according to the Illumina small RNA sample preparation guide vl .5 with the following exceptions. Custom 5' RNA ligation adapters were synthesized with a 4 nt nucleotide barcode sequence (Table 7). Small RNA libraries were pooled and sequenced on one sequencing lane of an Illumina GAII sequencer (Genome Technology Core, Whitehead Institute or NEB). Novocraft software
(www.novocraft.com) was used for sequence alignment. Reference genome was E. coli K12 substr. MG1655. We wrote Perl software scripts for data analysis. Original data and software scripts are available upon request.
[00170] rriRNA profiling by microarray and deep sequencing. siRNAs and pro-siRNAs (4 tiM) were transfected into HeLa-dlEGFP cells and RNA was isolated 24 hr post-transfection. Non- targeting siRNA #4 (Dharmacon) was used as negative control siRNA. Data from biological duplicates were analyzed at the Microarray Core, Dana Farber Cancer Institute for microarray analysis using GeneChip 1.0 ST (Affymetrix). Microarray data was analyzed using dChip software and p values of gene expression changes were calculated using paired T-test method34. Original data and analysis files are available upon request.
[00171] For RNA deep sequencing, Ribo-Zero rRNA Removal Kits (Epicentre) was used to remove large ribosomal RNAs from total RNA following the manufacturer's protocol. rRNA-depleted RNA (from 500 ng total RNA) was used to construct deep sequencing library using NEBNext Ultra RNA Library Prep Kit for Illumina (NEB #E7530) according to the manufacturer's protocol. Illumina GAII was used for sequencing (NEB). Tophat and Cufflinks software suites were used to analyzed the RNA deep sequencing data from biological duplicates. Reference genome was Human genome GRCh37/hgl9 and annotations of lincRNA transcripts were downloaded from UCSD genome browser. Original data and analysis files are available upon request.
[00172] Flow cytometry. For EGFP, cells were removed from plates by trypsin digestion and re- suspended in FACS buffer, DPBS (Invitrogen) containing 2% heat-inactivated FBS. Intracellular staining of p24 antigen was performed using an Intracellular Staining Kit (Invitrogen) according to the manufacturer's protocol and fluorescein-labeled p24 antibody (1 :200, Beckman Coulter, cat#KC57-FITC). Fluorescence was analyzed on a FACSCalibur (BD) using FlowJo software (Tree Star).
[00173] HIV infection and TZM-bl assay. HeLa-CD4 cells were transfected with 4 nM siRNA and pro-siRNA in 24 well plates ( 1 x 105 cells/well). Cells were infected 12 hr post-transfection with HIVIIIB (-400 ng/ml p24) and culture medium was changed 12 hr post-infection. For
HIVUG29 U87.CD4.CXCR4 cells were used and for HIVIN22 U87.CD4.CCR5 cells were used. Culture medium was collected for TZM-bl assay and RNA was extracted for qRT-PCR 24-36 hr post- infection. TZM-bl cells, plated in 24 well plates (l x lO5 cells/well) 12 hr before, were analyzed 24 later by luciferase assay performed using a Luciferase Assay System kit (Promega) following the manufacturer's protocol.
[00174] RNase A digestion assay for E. coli total RNA. ~2 ug of total E. coli RNA were incubated with 1.0 unit of RNase A for 15 min at 37°C in IX DNase I reaction buffer (NBE) supplemented with 400 mM NaCl. The resulting products were analyzed on a 0.8% agarose gel containing EtBr.
REFERENCES
1. Fire, A. et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391, 806-811 (1998).
2. Hamilton, A.J. & Baulcombe, D.C. A species of small antisense RNA in posttranscriptional gene silencing in plants. Science 286, 950-952 (1999).
3. Lejeune, E. & Allshire, R.C. Common ground: small RNA programming and chromatin modifications. Curr Opin Cell Biol 23, 258-265 (2011).
4. Elbashir, S.M. et al. Duplexes of 21 -nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature Al l, 494-498 (2001).
5. Caplen, N.J., Parrish, S., Imani, F., Fire, A. & Morgan, R.A. Specific inhibition of gene expression by small double-stranded RNAs in invertebrate and vertebrate systems. Proc Natl Acad Sci USA 98, 9742-9747 (2001).
6. Rettig, G.R. & Behlke, M.A. Progress toward in vivo use of siRNAs-II. Mol Ther 20, 483-512 (2012).
7. Timmons, L. & Fire, A. Specific interference by ingested dsRNA. Nature 395, 854 (1998).
8. Myers, J.W., Jones, J.T., Meyer, T. & Ferrell, J.E., Jr. Recombinant Dicer efficiently converts large dsRNAs into siRNAs suitable for gene silencing. Nat Biotechnol 21 , 324-328 (2003).
9. Yang, D. et al. Short RNA duplexes produced by hydrolysis with Escherichia coli RNase III mediate effective RNA interference in mammalian cells. Proc Natl Acad Sci U SA 99, 9942- 9947 (2002).
10. Morlighem, J.E., Petit, C. & Tzertzinis, G. Determination of silencing potency of synthetic and RNase Ill-generated siRNA using a secreted luciferase assay. Biotechniques 42, 599-605 (2007).
11. Semizarov, D. et al. Specificity of short interfering RNA determined through gene expression signatures. Proc Natl Acad Sci USA 100, 6347-6352 (2003).
12. Voinnet, O., Pinto, Y.M. & Baulcombe, D.C. Suppression of gene silencing: a general strategy used by diverse DNA and RNA viruses of plants. Proc Natl Acad Sci U S A 96, 14147-14152 (1999).
13. Silhavy, D. et al. A viral protein suppresses RNA silencing and binds silencing-generated, 21- to 25-nucleotide double-stranded RNAs. Embo J 21, 3070-3080 (2002). Vargason, J.M., Szittya, G., Burgyan, J. & Hall, T.M. Size selective recognition of siRNA by an RNA silencing suppressor. Cell 115, 799-811 (2003).
Jin, J., Cid, M., Poole, C.B. & McReynolds, L.A. Protein mediated miRNA detection and siRNA enrichment using pl9. Biotechniques 48, xvii-xxiii (2010).
Chu, M., Desvoyes, B., Turina, M., Noad, R. & Scholthof, H.B. Genetic dissection of tomato bushy stunt virus pl9-protein-mediated host-dependent symptom induction and systemic invasion. Virology 266, 79-87 (2000).
Knight, S.W. & Bass, B.L. A role for the RNase III enzyme DCR-1 in RNA interference and germ line development in Caenorhabditis elegans. Science 293, 2269-2271 (2001).
Timmons, L., Court, D.L. & Fire, A. Ingestion of bacterially expressed dsRNAs can produce specific and potent genetic interference in Caenorhabditis elegans. Gene 263, 103-112 (2001). Babitzke, P., Granger, L., Olszewski, J. & Kushner, S.R. Analysis of mRNA decay and rRNA processing in Escherichia coli multiple mutants carrying a deletion in RNase III. J Bacteriol 175, 229-239 (1993).
Cummins, J.M. et al. The colorectal microRNAome. Proc Natl Acad Sci U S A 103, 3687- 3692 (2006).
Jackson, A.L. et al. Position-specific chemical modification of siRNAs reduces "off-target" transcript silencing. Rna 12, 1197-1205 (2006).
Spankuch, B. et al. Cancer inhibition in nude mice after systemic application of U6 promoter- driven short hairpin RNAs against PLK1. J Natl Cancer Inst 96, 862-872 (2004).
Lee, S.K. et al. Lentiviral delivery of short hairpin RNAs protects CD4 T cells from multiple clades and primary isolates of HIV. Blood 106, 818-826 (2005).
Sugiyama, R., Habu, Y., Ohnari, A., Miyano-Kurosaki, N. & Takaku, H. RNA interference targeted to the conserved dimerization initiation site (DIS) of HIV-1 restricts virus escape mutation. J Biochem 146, 481-489 (2009).
Jayaprakash, A.D., Jabado, O., Brown, B.D. & Sachidanandam, R. Identification and remediation of biases in the activity of RNA ligases in small-RNA deep sequencing. Nucleic Acids Res 39, el41 (2011).
Jackson, A.L. et al. Expression profiling reveals off-target gene regulation by RNAi. Nat Biotechnol 2\, 635-637 (2003).
Weinberg, D.E., Nakanishi, K., Patel, D.J. & Bartel, D.P. The inside-out mechanism of Dicers from budding yeasts. Cell 146, 262-276 (2011).
Tenllado, F., Martinez-Garcia, B., Vargas, M. & Diaz-Ruiz, J.R. Crude extracts of bacterially expressed dsRNA can be used to protect plants against virus infections. BMC Biotechnol 3, 3 (2003).
Xiang, S., Fruehauf, J. & Li, C.J. Short hairpin RNA-expressing bacteria elicit RNA interference in mammals. Nat Biotechnol 24, 697-702 (2006). 30. Zhao, H.F. et al. High-throughput screening of effective siRNAs from RNAi libraries delivered via bacterial invasion. Nat Methods 2, 967-973 (2005).
31. Nakanishi, K., Weinberg, D.E., Bartel, D.P. & Patel, D.J. Structure of yeast Argonaute with guide RNA. Nature 486, 368-374 (2012).
32. Dancz, C.E., Haraga, A., Portnoy, D.A. & Higgins, D.E. Inducible control of virulence gene expression in Listeria monocytogenes: temporal requirement of listeriolysin O during intracellular infection. J Bacteriol 184, 5935-5945 (2002).
33. Pall, G.S. & Hamilton, A.J. Improved northern blot method for enhanced detection of small RNA. Nat Protoc 3, 1077-1084 (2008).
34. Li, C. & Wong, W.H. Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci U S A 98, 31-36 (2001).
Table 1. Gel clot Limulus amoebocyte lysate (LAL) endotoxin assays of HPLC-purified pro-siRNA
Gel clot LAL assay
Sample (limit of detection 0.25 EU/ml)
H20
LPS (4 ng/ml) +
P19 RNA SDS eluate (~100 nM) +
HPLC pro-siRNA 16 nM
HPLC pro-siRNA 64 nM
HPLC pro-siRNA 160 nM
HPLC pro-siRNA 320 nM
Table 2. Sequencing reads and alignment summary of RNA deep sequencing data
Percentage
Type Sample name Total reads Aligned reads aligned
Total RNA NC siRNA-1 21,954,641 19,032,496 86.7%
Total RNA NC siRNA-2 26,914,681 22,462,181 83.5%
Total RNA EGFP siRNA-1 25,659,586 21,237,241 82.8%
Total RNA EGFP siRNA-2 23,235,174 19,588,652 84.3%
Total RNA EGFP 100 pro-siRNA- 1 27,110,365 23,381,006 86.2%
Total RNA EGFP 100 pro-siRNA-2 22,690,638 19,433,997 85.6%
Total RNA EGFPFL pro-siRNA- 1 27,914,511 23,335,378 83.6%
Total RNA EGFPFL pro-siRNA-2 21,572,278 18,178,029 84.3% small RNA EGFPFL/EGFPFL- 1 3,291,738 3,119,677 94.8% small RNA EGFP 100 2,967,297 1,488,213 50.2% small RNA LMNA 1,659,890 1,382,441 83.3% small RNA TP53 5,446,487 4,462,318 81.9% small RNA PLK1 2,938,903 2,309,515 78.6% small RNA vif 1,869,202 1,493,137 79.9% small RNA gagB200 5,326,736 3,640,886 68.4% small RNA gagB500 7,168,829 5,017,221 70.0% small RNA EGFPFL -2 5,507,507 4,075,642 74.0% small RNA EGFP Hotspot-1 6,483,321 5,425,661 83.7% small RNA EGFP Hotspot-2 6,485,138 4,019,427 62.0%
Table 3. EGFPFL pro-siRNAs for testing strand bias
Figure imgf000052_0001
Table 4. List of E. coli strains
Name Genotype Source/reference
DH5a fhuA2A(argF-lacZ)U169 phoA NEB (C2987) glnV44 8O A(lacZ)M15 gyrA96 recAl
relAl endAl thi-l hsdR17
T7 Express Iq MiniF lacP(CamR) / ' fhuA2 lacZ::T7 genel NEB (C3016)
[Ion] ompT gal sulAl 1 R(mcr-73::miniTnlO- -Tets)2 [dcm] R(zgb-210::Tnl0-Te ) endAl
A ( mcrC-mrr) 114: :IS10
HT115(DE3) W3110 rnc-14::ATnJO XDE3 Timmons et al.
(2001), gift of Gary Ruvkun
BL21(DE3) fliuA2 [Ion] ompT gal (X DE3) [dcm] AhsdS NEB (C2527)
X DE3 = X sBamHIo AEcoRI-B
int: : (lad: :PlacUV5::T7 genel) i21 Anin5
SK7622 thyA715 Arnc-38::Kmr Babitzke et al.
(1993), gift of Sidney Kushner
MG1655 AlacZYA F- lambda- ilvG- rfb-50 rph-1 AlacZYA Gift from S. Garrity
MG1655 AlacZYA rncl 4 F- lambda- ilvG- rfb-50 rph-1 AlacZYA This study
Arncl4
MG1655 AlacZYA rnc38 F- lambda- ilvG- rfb-50 rph-1 AlacZYA This study
Arnc38 Table 5. List of plasmids
Name Purpose pcDNA3.1+ Empty control plasmid
pcDNA3.1- For expression of TREXl-FLAG protein directed from TREX1-FLAG the CMV promoter
pcDNA3.1-P19- For expression of P19-FLAG from the CMV promoter FLAG
pcDNA3.1-His- For expression of His-P19 from the CMV promoter P19
pcDNA3.1-P19- For expression of His-P19 3942 mutant from the CMV 3942-His promoter
pcDNA3.1-P19- For expression of His-P19 7172 mutant from the CMV 7172-His promoter
pcDNA3.1 -RNase For expression of FLAG tagged E. coli RNase III III
pRSF-GST-P19- For expression of GST-P19-His from T7 promoter His
pCDF-GST-P19- For expression of GST-P19-His from T7 promoter His
pCDF-RNase III For expression of FLAG tagged E. coli RNase III from
T7 promoter
L4440-EGFP For expression of double stranded eGFP RNA from convergent T7 promoters
pGEX-4T-l-P19- For expression of GST-P19-His from Tac promoter His
pGEX-4T-l-P19- For expression of GST-P19-His from Tac promoter, and T7 hairpin RNA from the T7 promoter
pGEX-4T-l-P19- For producing EGFPFL pro-siRNA
T7-EGFPFL
pGEX-4T-l-P19- For producing EGFP Hotspotl pro-siRNA
T7-EGFP-
Hotspotl
pGEX-4T-l-P19- For producing EGFP Hotspot2 pro-siRNA
T7-EGFP- Hotspot2
pGEX-4T-l-P19- For producing EGFP100 pro-siRNA
T7-EGFP100
pGEX-4T-l-P19- For producing LMNA pro-siRNA
T7-LMNA
pGEX-4T-l-P19- For producing PLK1 pro-siRNA
T7-PLK1
pGEX-4T-l-P19- For producing TP53 pro-siRNA
T7-TP53
pGEX-4T-l-P19- For producing HIV-vif pro-siRNA
T7-Vif
pGEX-4T-l-P19- For producing gagB200 pro-siRNA
T7-GagB200
pGEX-4T-l-P19- For producing gagB500 pro-siRNA
T7-GagB500
pLIV-1 Empty control plasmid
pLIV-l-His-P19 For expression of His-P19 protein in L. monocytogenes
Table 6. List of DNA oligonucleotides
Name Sequence (5'-3') SEQ ID Purpose
NO
PI 9-F-NheI AATCGCTAGCATGGAACGAGCTAT 10 pcDNA3.1-P19- ACAAGGA FLAG
PI 9-R-BamHI AATCGGATCCCTCGCTTTCTTTTTC 11 pcDNA3.1-P19- GAAGG FLAG
AATCGGATCCGATCCAAAAAAGAA 12 pcDNA3.1-P19-
GAGAAAGGTAGATCCAAAAAAGAA FLAG
GAGAAAGGTA
PI 9NLS-R AATCCTCGAGTCACTTATCGTCGTC 13 pcDNA3.1-P19- ATCCTTGTAATCGCCTACCTTTCTCT FLAG
AATCGCTAGCATGCACCACCACCA 14 pcDNA3.1-P19- CCACCACGCGGGCGAACGAGCTAT His
ACAAGGA P19-R-BamHI AATCGGATCCTCACTCGCTTTCTTT 15 pcDNA3.1-P19- TTCGAAGG His
P19W3942G-F CCGAGTGGCACTGAGGGCCGGCTA 16 pcDNA3.1-P19- CATAACGATGAGACGAATTC 3942-His
P 19 W3942G- TAGCCGGCCCTCAGTGCCACTCGGA 17 pcDNA3.1-P19- R CTTTCGTCAGGAAGTTTGA 3942-His
P 19KR7172A GTTGTATTTGCGGGCTATCTCAGAT pcDNA3.1-P19- G-F ACGACAGGACGGAAGCTTC 7172-His
P19KR7172A TCTGAGATAGCCCGCAAATACAAC 19 pcDNA3.1-P19- G-R TTTCCCGAAACCCCAGCTTT 7172-His
P19F-XbaI AATATCTAGAATGGAACGAGCTAT 20 pLIV-l -P19-His
ACAAGGA
P19R-His- AATCTCTAGATCAGTGGTGGTGGTG 21 pLIV-l -P19-His Xbal GTGGTG
PI 9-F-BamHI AATCGGATCCATGGAACGAGCTAT 22 pGEX-4T-l -P19- ACAAGGA His
AATCCTCGAGTCAGTGGTGGTGGTG 23 pGEX-4T-l -P19-
GTGGTGCTCGCTTTCTTTTTCGAAG His
G
ACTTGCTAGCATGGATTACAAGGAT 24 pcDNA3.1-RNase
GACGACGATAAGAACCCCATCGTA III and pCDF-
ATTAATCG RNase III rnc-BamHI-R ATCGGGATCCTCATTCCAGCTCCAG 25 RNase III and pCDF-RNase III
His-T7-Sacl ATCGAGCTCCCCTATAGTGAGTCGT 26 pGEX-4T-l -P19- ATTAGATTCAGTGGTGGTGGTGGTG T7
GT
Linker3-F ATGAATTCGTCGACACTGCGGCCGC 27 pGEX-4T-l -P19- TCTAGAGGGCCCGTTTAAACCCGCT T7
ATCTCGAGAATGAGCTCGCTGATCA 28 pGEX-4T-l -P19-
GCGGGTTTAAACGGGCCCTCTAGA T7
G
GST-F-Ndel ATCCCATATGTCCCCTATACTAGGT 29 pRSF-GST-P19- TATTG His, pCDF-GST- P19-His
His-R-Xhol AATCCTCGAGTCAGTGGTGGTGGTG 30 pRSF-GST-P19- GTGGTG His, pCDF-GST- P19-His EGFP-F-SacI AATCGAGCTCCATGGTGAGCAAGG 31 pGEX-4T-l -P19- GCGAGGA T7-EGFPFL
EGFP-F-NotI AATCGCGGCCGCATGGTGAGCAAG 32 pGEX-4T-l -P19- GGCGAGGA T7-EGFPFL
EGFP-R-Sall AATCGTCGACCTACTTGTACAGCTC 33 pGEX-4T-l -P19- GTCCA T7-EGFPFL
EGFP-F-XhoI AATCCTCGAGCTACTTGTACAGCTC 34 pGEX-4T-l -P19- GTCCA T7-EGFPFL,
EGFP northern blot probe
EGFPHS1 -F- ATCCGCGGCCGCATGGTGAGCAAG 35 pGEX-4T-l -P19- Notl GGCGAGGAG T7-EGFP-
Hotspotl
EGFPHS1 -F- ATCGAGCTCATGGTGAGCAAGGGC 36 pGEX-4T-l -P19- Sad GAGGAG T7-EGFP-
Hotspotl
EGFPHS1 -R- ATCGTCGACCAGGGTGTCGCCCTCG 37 pGEX-4T-l -P19- Sail AACTT T7-EGFP-
Hotspotl
EGFPHS1 -R- ATCCTCGAGCAGGGTGTCGCCCTCG 38 pGEX-4T-l -P19- Xhol AACTT T7-EGFP-
Hotspotl
EGFPHS2-F- ATCCGCGGCCGCGTGAACCGCATC 39 pGEX-4T-l -P19- Notl GAGCTGAAG T7-EGFP-
Hotspot2
EGFPHS2-F- ATCGAGCTCGTGAACCGCATCGAG 40 pGEX-4T-l -P19- Sad CTGAAG T7-EGFP-
Hotspot2
EGFPHS2-R- ATCGTCGACCTACTTGTACAGCTCG 41 pGEX-4T-l -P19- Sail TCCAT T7-EGFP-
Hotspot2
EGFPHS2-R- ATCCTCGAGCTACTTGTACAGCTCG 42 pGEX-4T-l -P19- Xhol TCCAT T7-EGFP-
Hotspot2
EGFP 100-F- AATCGAGCTCCCGCTACCCCGACCA 43 pGEX-4T-l -P19- Sad CATGAA T7-EGFP100
EGFP 100-F- AATCCGCGGCCGCCCGCTACCCCG 44 pGEX-4T-l -P19- Notl ACCACATGAA T7-EGFP100
EGFP 100-R- AATCGTCGACGTTGCCGTCGTCCTT 45 pGEX-4T-l -P19- Sail GAAGAA T7-EGFP100 EGFP 100-R- AATCCTCGAGGTTGCCGTCGTCCTT 46 pGEX-4T-l -P19- Xhol GAAGAA T7-EGFP100
TP53-R-SalI AATCGTCGACCAACCTCAGGCGGC 47 pGEX-4T-l -P19- TCATAGG T7-TP53
TP53-RXhoI AATCCTCGAGCAACCTCAGGCGGC 48 pGEX-4T-l -P19- TCATAGG T7-TP53
TP53-F-Not AATCGCGGCCGCTACTCCCCTGCCC 49 pGEX-4T-l -P19- TCAACAAGATG T7-TP53
TP53-F-SacI AATCGAGCTCTACTCCCCTGCCCTC 50 pGEX-4T-l -P19- AACAAGATG T7-TP53
HIV-Vif-F- AATCGAGCTCGGAAAACAGATGGC 51 pGEX-4T-l -P19- Sad AGGTGATG T7-Vif
HIV-Vif-F- AATCGCGGCCGCGGAAAACAGATG 52 pGEX-4T-l -P19- Notl GCAGGTGATG T7-Vif
HIV-Vif-R- AATCGTCGACCTAGTGTCCATTCAT 53 pGEX-4T-l -P19- Sail TGTGTGG T7-Vif
HIV-Vif-R- AATCCTCGAGCTAGTGTCCATTCAT 54 pGEX-4T-l -P19- Xhol TGTGTGG T7-Vif
LaminAC-F- AATCGAGCTCCAAGACCCTTGACTC 55 pGEX-4T-l -P19- Sad AGTAGCC T7-LMNA
LaminAC-F- AATCGCGGCCGCCAAGACCCTTGA 56 pGEX-4T-l -P19- Notl CTCAGTAGCC T7-LMNA
LaminAC-R- AATCGTCGACCAGCTCCTTCTTATA 57 pGEX-4T-l -P19- Sail CTGCTCCA T7-LMNA
LaminAC-R- AATCCTCGAGCAGCTCCTTCTTATA 58 pGEX-4T-l -P19- Xhol CTGCTCCA T7-LMNA
PLK1 -F-Notl AATCGCGGCCGCTCTCTGCTGCTCA 59 pGEX-4T-l -P19- AGCCGCAC T7-PLK1
PLK1 -F-Sacl AATCGAGCTCTCTCTGCTGCTCAAG 60 pGEX-4T-l -P19- CCGCAC T7-PLK1
PLKl-R-Sall AATCGTCGACAAGTCTCAAAAGGT 61 pGEX-4T-l -P19- GGTTTGCC T7-PLK1
PLKl-R-XhoI AATCCTCGAGAAGTCTCAAAAGGT 62 pGEX-4T-l -P19- GGTTTGCC T7-PLK1
Gag200-FNotI ATCCGCGGCCGCTGTGGCAAAGAA pGEX-4T-l -P19- GGGCACACAG T7-GagB200
Gag200-FSacI ATCGAGCTCTGTGGCAAAGAAGGG pGEX-4T-l -P19- CACACAG T7-GagB200
Gag200-RSalI ATCGTCGACTCTTCTGGTGGGGCTG 65 pGEX-4T-l -P19- TTGGCT T7-GagB200
Gag200- ATCCTCGAGTCTTCTGGTGGGGCTG 66 pGEX-4T-l -P19- RXhoI TTGGCT T7-GagB200
Gag500-FNotI ATCCGCGGCCGCAAGCATTGGGAC 67 pGEX-4T-l -P19- CAGCGGCTAC T7-GagB500 Gag500-FSacI ATCGAGCTCAAGCATTGGGACCAG 68 pGEX-4T-l -P19- CGGCTAC T7-GagB500 Gag500-RSalI ATCGTCGACTTATTGTGACGAGGGG 69 pGEX-4T-l -P19- TCGTTG T7-GagB500
Gag500- ATCCTCGAGTTATTGTGACGAGGGG 70 pGEX-4T-l -P19- RXhoI TCGTTG T7-GagB500
SiSEQl CAAGCAGAAGACGGCATACGA 71 Deep sequencing library PCR
SiSEQ2 AATGATACGGCGACCACCGACAGG 72 Deep sequencing
TTCAGAGTTCTACAGTCCGA library PCR
GAPDH For CTGGGCTACACTGAGCACC 73
AAGTGGTCGTTGAGGGCAATG 126
GAPDH Rev
IL12 For CACTCCCAAAACCTGCTGCTGAG 74 qRT-PCR IL12 Rev TCTCTTCAGAAGTGCAAGGGTA 75 qRT-PCR IL6 For GATGAGTACAAAAGTCCTGATCCA 76 qRT-PCR IL6 Rev CTGCAGCCACTGGTTCTGT 77 qRT-PCR IL8 For AGACAGCAGAGCACACAAGC 78 qRT-PCR IL8 Rev ATGGTTCCTTCCGGTGGT 79 qRT-PCR TNFA For CAGCCTCTTCTCCTTCCTGAT 80 qRT-PCR TNFA Rev GCCAGAGGGCTGATTAGAGA 81 qRT-PCR Vif For AGGGAAAGCTAGGGGATGGTTTT 82 qRT-PCR Vif Rev CCCAAATGCCAGTCTCTTTCTCC 83 qRT-PCR TN22-Vif For AAAGAGAGCTAATGGATGGTTTT 84 qRT-PCR IN22-Vif Rev CCCAAATGCCAATCTCTTTCCCC 85 qRT-PCR UG29-Vif For AAAGAAAGCTACTGGTTGGTGTT 86 qRT-PCR UG29-vif Rev qRT-PCR GagABC For CCTAGGAAAAAGGGCTGTTGGA 88 qRT-PCR GagABC Rev AGGAAGGCCAGATCTTCCCTAAA 89 qRT-PCR IFITlFor GCCACAAAAAATCACAAGCCA 90 qRT-PCR IFITlRev CCATTGTCTGGATTTAAGCGG 91 qRT-PCR LMNA For AGCAGCGTGAGTTTGAGAGC 92 qRT-PCR LMNA Rev CCAGCTTGGCAGAATAAGTCTT 93 qRT-PCR PLK1 For CGAGGACAACGACTTCGTGTT 94 qRT-PCR PLK1 Rev ACAATTTGCCGTAGGTAGTATCG 95 qRT-PCR TP53 For ACAGCTTTGAGGTGCGTGTTT 96 qRT-PCR TP53 Rev CCCTTTCTTGCGGAGATTCTCT 97 qRT-PCR eGFP For ACGTAAACGGCCACAAGTTC 98 qRT-PCR eGFP Rev AAGTCGTGCTGCTTCATGTG 99 qRT-PCR EGFP-T7-F ACTAATACGACTCACTATAGGGAT 100 EGFP northern
GGTGAGCAAGGGCGAGGA blot probe
EGFPFL-sil-F CGTGCTGCTGCCCGACAACCACTAC 101 Solution
CT hybridization
EGFPFL-sil-R GAGGTAGTGGTTGTCGGGCAGCAG 102 Solution
CACG hybridization
EGFPFL-si2-F CTACAACAGCCACAACGTCTATATC 103 Solution
A hybridization
EGFPFL-si2-R TGATATAGACGTTGTGGCTGTTGTA 104 Solution
G hybridization
EGFPFL-si3-F CCTGGTCGAGCTGGACGGCGACGT 105 Solution
AA hybridization
EGFPFL-si3-R TTACGTCGCCGTCCAGCTCGACCAG 106 Solution
G hybridization
ACH-5 TATGAGGAACAGATTTTCTCACATG 107 Control oligo for
G solution
hybridization Table 7. RNA adapters for small RNA deep sequencing libraries
Name Sequence (5'-3') SEQ ID Purpose
NO
5ADPT-2 GUUCAGAGUUCUACAGUCCGACGAUCGCUU 108 5' adapter
EGFPFL
5ADPT-3 GUUCAGAGUUCUACAGUCCGACGAUCGAGU 109 5' adapter
EFGP100
5ADPT-5 GUUCAGAGUUCUACAGUCCGACGAUCCGUU 110 5' adapter for
PLK1
5ADPT-6 GUUCAGAGUUCUACAGUCCGACGAUCCCGU 111 5' adapter for
LMNA
5ADPT-7 GUUCAGAGUUCUACAGUCCGACGAUCCACU 112 5' adapter
HlV-Vif
3ADPT UCGUAUGCCGUCUUCUGCUUGUidT 3' adapter all libraries

Claims

What is claimed herein is:
1. A bacterial cell comprising a siRNA-binding polypeptide and a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
2. The bacterial cell of claim 1, wherein the siRNA-binding polypeptide comprises a purification tag.
3. The bacterial cell of any of claims 1-2, wherein the siRNA-binding polypeptide is encoded by a nucleic acid.
4. The bacterial cell of any of claims 1-3, wherein the siRNA-binding polypeptide is selected from the group consisting of:
pi 9 polypeptide; tombusvirus pi 9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
5. The bacterial cell of any of claims 1-4, wherein the dsRNA is greater than 21 nucleotides in length.
6. The bacterial cell of any of claims 1-5, wherein the dsRNA is a hairpin RNA.
7. The bacterial cell of any of claims 1-6, wherein the bacterial cell expresses an RNase III polypeptide.
8. The bacterial cell of any of claims 1-7, wherein the bacterial cell expresses an RNase III polypeptide encoded by an exogenous nucleic acid sequence.
9. The bacterial cell of any of claims 1-8, wherein the bacterial cell is an Escherichia coli cell.
10. The bacterial cell of any of claims 1-9, wherein at least one of the siRNA-binding polypeptide and the dsRNA are constitutively expressed.
11. The bacterial cell of any of claims 1-10, wherein at least one of the siRNA-binding
polypeptide and the dsRNA are inducibly expressed.
12. The bacterial cell of any of claims 1-11, wherein the DNA encoding at least one of the
siRNA-binding polypeptide or the dsRNA is part of a plasmid.
13. The bacterial cell of any of claims 1-12, wherein the dsRNA comprises nucleic acid
sequences substantially complementary to a multiplicity of target RNAs.
14. A method of producing one or more siRNA species which can inhibit the expression of a target RNA, the method comprising:
culturing a bacterial cell of any of claims 1-13 under conditions suitable for the production of siRNAs.
15. The method of claim 14, further comprising a second step of isolating the siRNA-binding polypeptide and eluting the siRNAs bound to the siRNA-binding polypeptide.
16. The method of any of claims 14-15, further comprising purifying the siRNAs eluted from the siRNA-binding polypeptide by HPLC.
17. The method of any of claims 14-16, further comprising contacting the cell with one or more modified nucleotides before or during the culturing step.
18. A pharmaceutical composition comprising a siRNA produced according to the method of any of claims 14-17.
19. The composition of claim 18, further comprising a population of siRNA species.
20. A pharmaceutical composition comprising a siRNA isolated from a bacterial cell of any of claims 1-13.
21. The composition of claim 20, further comprising a population of siRNA species.
22. The use of a siRNA produced according to the method of any of claims 14-17 in the
production of a medicament.
23. The use of a siRNA isolated from a bacterial cell of any of claims 1 -13 in the production of a medicament.
24. A vector comprising;
a nucleic acid encoding a siRNA-binding polypeptide; and
a dsRNA cloning site.
25. The vector of claim 24, wherein the dsRNA cloning site comprises at least one restriction enzyme site and can accept the insertion of at least one nucleic acid sequence such that a dsRNA is encoded and can be expressed.
26. A vector comprising:
a nucleic acid encoding a siRNA-binding polypeptide; and
a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
27. The vector of any of claims 24-26, wherein the siRNA-binding polypeptide is selected from the group consisting of:
pi 9 polypeptide; tombusvirus pi 9 polypeptide; B2 polypeptide; HC-Pro polypeptide; p38 polypeptide; pl22 polypeptide; pl30 polypeptide; p21 polypeptide; plb polypeptide; and NS3 polypeptide.
28. The vector of any of claims 24-27, wherein the vector is a plasmid.
29. The vector of claim 28, wherein the plasmid further comprises a bacterial origin of
replication.
30. A library of siRNA species, the library comprising:
a plurality of clonal bacterial cell populations;
wherein each clonal population is comprises bacterial cells of any of claims 1-13.
31. A library of siRNA species, the library comprising:
a plurality of populations of siRNAs; wherein each population of siRNAs is obtained according to the methods of any of claims 14-17.
32. The library of claim 31, wherein each population of siRNAs binds to a single target RNA.
33. A kit comprising a bacterial cell of any of claims 1-13.
34. A kit for the production of one or more species of siRNA, the kit comprising;
a bacterial cell comprising an siRNA-binding polypeptide; and
at least one vector comprising a dsRNA cloning site.
35. A kit for the production of one or more species of siRNA, the kit comprising:
a bacterial cell comprising an siRNA-binding polypeptide; and
at least one vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
36. A kit comprising the vector of any of claims 24-29.
37. A kit for the production of one or more species of siRNA, the kit comprising two vectors;
wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and
wherein the second vector comprises a dsRNA cloning site.
38. A kit for the production of one or more species of siRNA, the kit comprising two plasmids;
wherein the first vector comprises a nucleic acid encoding a siRNA-binding polypeptide; and
wherein the second vector comprises a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
39. The kit of any of claims 33-38, wherein at least one vector is a plasmid.
40. The kit of claim 39, wherein the plasmid further comprises a bacterial origin of replication.
41. The kit of any of claims 33-40, wherein the kit further comprises a bacterial cell.
42. A kit for the production of one or more species of siRNA, the kit comprising;
a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA cloning site.
43. A kit for the production of one or more species of siRNA, the kit comprising;
a bacterial cell comprising a nucleic acid encoding a siRNA-binding polypeptide; and a vector comprising a dsRNA comprising a nucleic acid sequence substantially complementary to a target RNA.
44. The kit of any of claims 33-43, wherein the siRNA-binding polypeptide comprises a
purification tag.
45. The kit of any of claims 33-44, wherein the siRNA-binding polypeptide is encoded by a nucleic acid.
46. The kit of any of claims 41-45, wherein the bacterial cell expresses an RNase III polypeptide.
47. The kit of any of claims 41-46, wherein the cell is an Escherichia coli cell.
48. The kit of any of claims 33-47, wherein at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to a constitutive promoter.
49. The kit of any of claims 33-48, wherein at least one of the siRNA-binding polypeptide and the dsRNA are operably linked to an inducible promoter.
50. The kit of any of claims 33-49, wherein the DNA encoding at least one of the siRNA-binding polypeptide or the dsRNA is part of a plasmid.
51. A kit comprising the library of any of claims 30-32.
PCT/US2014/010784 2013-01-11 2014-01-09 Methods and compositions for the production of sirnas WO2014110205A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/758,924 US9840703B2 (en) 2013-01-11 2014-01-09 Methods and compositions for the production of siRNAs
US15/804,258 US10508276B2 (en) 2013-01-11 2017-11-06 Methods and compositions for the production of siRNAs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361751489P 2013-01-11 2013-01-11
US61/751,489 2013-01-11

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/758,924 A-371-Of-International US9840703B2 (en) 2013-01-11 2014-01-09 Methods and compositions for the production of siRNAs
US15/804,258 Continuation US10508276B2 (en) 2013-01-11 2017-11-06 Methods and compositions for the production of siRNAs

Publications (1)

Publication Number Publication Date
WO2014110205A1 true WO2014110205A1 (en) 2014-07-17

Family

ID=51167361

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/010784 WO2014110205A1 (en) 2013-01-11 2014-01-09 Methods and compositions for the production of sirnas

Country Status (2)

Country Link
US (2) US9840703B2 (en)
WO (1) WO2014110205A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112585260A (en) * 2018-03-31 2021-03-30 美国卵石实验室公司 Systems, methods, and compositions for producing sRNA using RNase III mutants to control host pathogen infection
WO2024046452A1 (en) * 2022-09-02 2024-03-07 Xiaomo Biotech Limited Inclusion body mediated method for double-stranded rna producing

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6843065B2 (en) 2015-03-30 2021-03-17 グリーンライト バイオサイエンシーズ インコーポレーテッドGreenlight Biosciences,Inc. Cell-free production of ribonucleic acid
CR20180525A (en) * 2016-04-06 2019-02-14 Greenlight Biosciences Inc RIBONUCLEIC ACID PRODUCTION FREE OF CELLS
JP2019531078A (en) * 2016-10-05 2019-10-31 シンジェンタ パーティシペーションズ アーゲー Method for preserving the biological activity of ribonucleic acid
US11193124B2 (en) * 2016-11-15 2021-12-07 City University Of Hong Kong Small-interfering RNA expression systems for production of small-interfering RNAs and their use
WO2022271972A1 (en) * 2021-06-23 2022-12-29 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing filamin a (flna)
CA3225740A1 (en) * 2021-06-29 2023-01-05 Alnylam Pharmaceuticals, Inc. Leucine-rich repeat kinase 2 (lrrk2) irna agent compositions and methods of use thereof
WO2024045077A1 (en) * 2022-08-31 2024-03-07 Xiaomo Biotech Limited A method for rnai fungicide generating and the rnai fungicide thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004011647A1 (en) * 2002-07-26 2004-02-05 Chiron Corporation Modified small interfering rna molecules and methods of use
US20060239971A1 (en) * 2003-02-21 2006-10-26 Mohapatra Shyam S Vectors for regulating gene expression
WO2007102140A2 (en) * 2006-03-08 2007-09-13 The Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin A semliki forest virus replication competent vector with enhanced biosafety
US20100209933A1 (en) * 2007-10-29 2010-08-19 Mcreynolds Larry A Methods and Compositions for Detection and Enrichment of Target Small RNAs

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7655790B2 (en) * 2002-07-12 2010-02-02 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
WO2007095152A2 (en) 2006-02-10 2007-08-23 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF sIRNA BY dsRNA BINDING DOMAIN FUSIONS TO PTD/CPPS

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004011647A1 (en) * 2002-07-26 2004-02-05 Chiron Corporation Modified small interfering rna molecules and methods of use
US20060239971A1 (en) * 2003-02-21 2006-10-26 Mohapatra Shyam S Vectors for regulating gene expression
WO2007102140A2 (en) * 2006-03-08 2007-09-13 The Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin A semliki forest virus replication competent vector with enhanced biosafety
US20100209933A1 (en) * 2007-10-29 2010-08-19 Mcreynolds Larry A Methods and Compositions for Detection and Enrichment of Target Small RNAs

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HUANG, LINFENG ET AL.: "Production of highly potent recombinant siRNAs in Escherichia coli", NAT. PROTOC., vol. 8, no. 12, 31 October 2013 (2013-10-31), pages 2325 - 2336 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112585260A (en) * 2018-03-31 2021-03-30 美国卵石实验室公司 Systems, methods, and compositions for producing sRNA using RNase III mutants to control host pathogen infection
EP3775188A4 (en) * 2018-03-31 2022-02-09 Pebble Labs USA Inc. Systems, methods and composition of using rnase iii mutants to produce srna to control host pathogen infection
US11800872B2 (en) 2018-03-31 2023-10-31 Pebble Labs Inc. Systems, methods and composition of using RNase III mutants to produce sRNA to control host pathogen infection
WO2024046452A1 (en) * 2022-09-02 2024-03-07 Xiaomo Biotech Limited Inclusion body mediated method for double-stranded rna producing

Also Published As

Publication number Publication date
US9840703B2 (en) 2017-12-12
US10508276B2 (en) 2019-12-17
US20150337306A1 (en) 2015-11-26
US20180273950A1 (en) 2018-09-27

Similar Documents

Publication Publication Date Title
US10508276B2 (en) Methods and compositions for the production of siRNAs
JP7412586B2 (en) VI-E and VI-F CRISPR-Cas systems and their use
Hasler et al. The lupus autoantigen La prevents mis-channeling of tRNA fragments into the human microRNA pathway
JP4339852B2 (en) Methods and compositions for gene silencing
Bouhouche et al. Functional specialization of Piwi proteins in Paramecium tetraurelia from post-transcriptional gene silencing to genome remodelling
Huang et al. Efficient and specific gene knockdown by small interfering RNAs produced in bacteria
CA3026110A1 (en) Novel crispr enzymes and systems
Zhang et al. RISC-interacting clearing 3’-5’exoribonucleases (RICEs) degrade uridylated cleavage fragments to maintain functional RISC in Arabidopsis thaliana
WO2017015101A1 (en) Methods for maximizing the efficiency of targeted gene correction
CN109415729B (en) Gene editing reagents with reduced toxicity
KR101605932B1 (en) Organic compositions to treat hsf1-related diseases
CA2989834A1 (en) Crispr enzymes and systems
WO2008140126A1 (en) Single-stranded cyclic rna, and method for production thereof
Lv et al. lncMGPF is a novel positive regulator of muscle growth and regeneration
CA3012607A1 (en) Crispr enzymes and systems
CN113906138A (en) Introduction of silencing activity into multiple functionally deregulated RNA molecules and modification of their specificity for a gene of interest
TW201402813A (en) Bi-functional short-hairpin RNA (bi-shRNA) specific for single-nucleotide KRAS mutations
WO2022068912A1 (en) Engineered crispr/cas13 system and uses thereof
US8846895B2 (en) Double-stranded RNA molecules with stability in mammalian body fluid, preparation and application thereof
IL302243A (en) Reverse transcription of polynucleotides comprising unnatural nucleotides
Huang et al. Production of highly potent recombinant siRNAs in Escherichia coli
KR20050026384A (en) Expression systems for stem loop rna molecule having rnai effect
WO2023274226A1 (en) Crispr/cas system and uses thereof
US20220389398A1 (en) Engineered crispr/cas13 system and uses thereof
WO2022188039A1 (en) Engineered crispr/cas13 system and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14738149

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14758924

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14738149

Country of ref document: EP

Kind code of ref document: A1